2005-Morphological and Functional Plasticity of Olfactory Ensheathing Cells

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 17

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/7395985

Morphological and functional plasticity of olfactory ensheathing cells

Article in Journal of Neurocytology · April 2005


DOI: 10.1007/s11068-005-5048-6 · Source: PubMed

CITATIONS READS

96 757

3 authors:

Adele J Vincent Adrian K West


University of Tasmania University of Tasmania
20 PUBLICATIONS 941 CITATIONS 121 PUBLICATIONS 4,648 CITATIONS

SEE PROFILE SEE PROFILE

Meng Inn Chuah


University of Tasmania
81 PUBLICATIONS 3,360 CITATIONS

SEE PROFILE

All content following this page was uploaded by Adele J Vincent on 24 May 2014.

The user has requested enhancement of the downloaded file.


Journal of Neurocytology 34, 65–80 (2005)

Morphological and functional plasticity of olfactory


ensheathing cells
A D E L E J . V I N C E N T 1 , A D R I A N K . W E S T 2 a n d M E N G I N N C H U A H 1, ∗
1
NeuroRepair Group, Discipline of Anatomy and Physiology, Private Bag 24, University of Tasmania Hobart, Tasmania, Australia 7001;
2
NeuroRepair Group, Discipline of Biochemistry, Private Bag 58, University of Tasmania, Hobart, Tasmania, Australia 7001
Inn.Chuah@utas.edu.au

Received 2 January 2005; revised 4 April 20005 ; accepted 8 July 2005

Abstract
In the primary olfactory pathway, olfactory ensheathing cells (OECs) extend processes to envelop bundles of olfactory axons
as they course towards their termination in the olfactory bulb. The expression of growth-promoting adhesion and extracellular
matrix molecules by OECs, and their spatially close association with olfactory axons are consistent with OECs being involved
in promoting and guiding olfactory axon growth. Because of this, OECs have been employed as a possible tool for inducing
axonal regeneration in the injured adult CNS, resulting in significant functional recovery in some animal models and promising
outcomes from early clinical applications. However, fundamental aspects of OEC biology remain unclear. This brief review
discusses some of the experimental data that have resulted in conflicting views with regard to the identity of OECs. We present
here recent findings which support the notion of OECs as a single but malleable phenotype which demonstrate extensive
morphological and functional plasticity depending on the environmental stimuli. The review includes a discussion of the
normal functional role of OECs in the developing primary olfactory pathway as well as their interaction with regenerating
axons and reactive astrocytes in the novel environment of the injured CNS. The use of OECs to induce repair in the injured
nervous system reflects the functional plasticity of these cells. Finally, we will explore the possibility that recent microarray
data could point to OECs assuming an innate immune function or playing a role in modulating neuroinflammation.

Introduction
The primary olfactory pathway consists of olfactory assume astrocyte (AC)-like functions in the CNS as ex-
neurons in the nasal cavity whose axons project through emplified by their formation of the glia limitans in the
the cribriform plate into the CNS to synapse with mitral olfactory bulb (Doucette, 1991); thus it was proposed
and tufted cells in the olfactory bulb. Along the course that they could belong to the astrocyte family. Even so,
of the unmyelinated olfactory nerves, they are accom- OECs were given an independent name, ‘ensheathing
panied by glial cells, widely known as olfactory en- cells’, in recognition of their potential to form a new
sheathing cells (OECs), which extend cytoplasmic pro- class of glia (Doucette, 1990). The notion of OECs con-
cesses to compartmentalise the small olfactory axons stituting a distinct class of glia has been reinforced in re-
(0.1–0.5 µm) into fascicles (Doucette et al., 1983). cent years in in vivo regeneration studies (Lakatos et al.,
The designation of OECs into a preexisting class of 2003) and tissue culture (Lakatos et al., 2000) in which
glial cell has been a contentious issue since their discov- they have demonstrated clear differences with SCs in
ery (Barnett, 2004; Barnett & Chang, 2004; Boyd et al., terms of cellular interaction with astrocytes and CNS
2003; Doucette & Devon, 1993; Raisman, 2001; Ramón- tissues.
Cueto & Valverde, 1995; Wewetzer et al., 2002). This dis- In recent years, debate has extended to the question
pute stems partly from the lack of a definitive molecu- of whether OECs should be classified as belonging ei-
lar identity for OECs. Initially OECs were called olfac- ther to a single population or to two subpopulations
tory nerve Schwann cells (SCs), suggesting that save based on their morphology and antigenic profile. This
for their location, OECs possess properties typical of brief review will discuss findings from various in vivo
peripheral nerve glia (Barber & Lindsay, 1982; Chuah and in vitro studies which have contributed to this de-
& Au, 1991; Cuschieri & Bannister, 1975; Gong et al., bate, including the morphological plasticity of OECs.
1994). However, it soon became apparent that OECs This is followed by a brief discussion of the normal

∗ To whom correspondence should be addressed.

0300–4864 
C 2005 Springer Science + Business Media, Inc.
66 V I N C E N T, W E S T and C H U A H

functional role of OECs with regard to developing olfac- the peripheral olfactory nerves and the olfactory nerve
tory nerves, and their physical interaction with regener- layer (ONL) of the bulb, express a number of antigens
ating axons and reactive astrocytes in the injured CNS. that can be detected immunohistochemically (Fig. 1;
Lastly we will raise the possibility that OECs could have note that for the purpose of this review, the following
other possible functional roles not normally associated discussion will only cover some of the molecules listed
with these cells, e.g. as a member of the innate immune in this figure). For example, research has demonstrated
system. the differential expression of OEC markers in the ONL.
The general consensus appears to be that all OECs ex-
press S100β and weak GFAP (and until recently, O4),
Antigenic characteristics of OECs in vivo
while (i) those in the outer ONL are weakly positive for
Early suggestions that antigenically distinct subpopu- the low-affinity neurotrophin receptor p75NTR and the
lations of OECs exist in the olfactory system were based embryonic neural cell adhesion molecule (E-NCAM),
on the variable intensity of immunohistochemical stain- and (ii) those in the inner ONL are neuropeptide Y
ing of the structural protein glial fibrillary acidic protein (NPY)-positive, but lack p75NTR and E-NCAM (Astic
(GFAP) in peripheral olfactory nerves (Barber & Lind- et al., 1998; Barnett et al., 1993; Franceschini & Barnett,
say, 1982; Pixley, 1992). It is now clear that OECs, both in 1996; Ubink et al., 1994; Valverde et al., 1992).

Fig. 1. A diagram representing the adult rat primary olfactory system. Olfactory ensheathing cells (OEC, pink areas) ensheath
bundles of olfactory receptor axons (olfactory nerves, ON) along their course through the lamina propria in the PNS. Olfactory
nerves and their accompanying OECs cross through the cribriform plate of the skull into the CNS and surround the olfactory
bulb to form the olfactory nerve layer. The olfactory nerve layer is comprised of outer and inner layers which represent zones
of axon-sorting and molecular expression by OECs. Axons then extend unaccompanied by OECs into glomeruli to synapse on
target cells. Olfactory ensheathing cells also contribute to the formation of the glial limitans (GL). Listed below are some of the
molecules produced by OECs in the lamina propria and olfactory nerve layer. BG, Bowman’s glands; BV, blood vessel; ORN,
olfactory receptor neuron.
Morphological and functional plasticity of olfactory ensheathing cells 67

S100β is one of a family of calcium-binding pro- freshly dissociated culture because of O4-positive ax-
teins which are involved in the regulation of numer- onal membrane fragments which adhere to the surface
ous intracellular processes including protein phospho- of OECs and are gradually phagocytosed (Wewetzer
rylation, proliferation and differentiation, and is ex- et al., 2005).
pressed widely being present in SCs, ACs and oligo- Neuropeptide Y displays the third expression pattern
dendrocytes (Donato, 1999). GFAP belongs to the class of OEC markers. It is thought to play a role in olfactory
of intermediate-sized cytoskeletal proteins and is com- neuronal differentiation or axonal growth because it is
monly accepted as a marker for astrocytes. In rats expressed in OECs in the developing ONL at E15 prior
S100β and GFAP appear first in the peripheral olfac- to the emergence of other laminae in the olfactory bulb
tory nerves at E14 and E15 respectively, and advance to (Ubink & Hökfelt, 2000). In adulthood it clearly delin-
the ONL just prior to birth (Astic et al., 1998; Valverde eates the inner ONL from the glomerular layer which
et al., 1992). A gradient also exists between the inner does not stain for NPY, with OECs in the peripheral
and outer sublaminae of the ONL; both markers are part of the ONL showing only weak staining (Au et al.,
restricted initially to the outer ONL and appear later 2002; Ubink et al., 1994; Ubink & Hökfelt, 2000).
in the inner ONL, around birth for S100β and increas- Because of the apparent difference in staining pat-
ing slowly from P7 to P20 for GFAP (Astic et al., 1998; tern for NPY, E-NCAM and p75NTR between the inner
Valverde et al., 1992). This delay in glial maturation and outer layers, it was thought that two subpopula-
may allow prolonged neuroplasticity during the de- tions of OECs exist in the adult ONL (Au et al., 2002;
velopmental period of major innervation of the bulb, Barnett & Chang, 2004; Franceschini & Barnett, 1996;
a phenomenon also observed in AC precursors (As- Ubink & Hökfelt, 2000). However, the exact definition
tic et al., 1998). The adult expression pattern of these of these subpopulations is questionable due to variabil-
markers is generally thought to be uniform throughout ity among studies resulting from factors such as, differ-
the ONL, with more intense immunostaining for S100β ential regulation of antigenic expression during devel-
than GFAP (Au et al., 2002; Franceschini & Barnett, 1996; opment and in adulthood, the use of different strains
Ubink et al., 1994). However, some of this GFAP stain- or species of organisms and a variety of antisera, and
ing may be due to the presence of interfascicular ACs the difficulty in identifying labelled structures by light
(Barnett et al., 1993) which infiltrate the ONL during microscopy. This latter problem is especially difficult
development (Doucette, 1984) or it may be due to the when immunostaining is detected in olfactory nerves
extended processes of periglomerular ACs (Astic et al., in which molecules may be expressed by both OECs
1998; Treloar et al., 1999). or olfactory axons, such as for the well-established
The expression of p75NTR , E-NCAM and O4 is promi- OEC markers E-NCAM, p75NTR and O4. Although re-
nent in OECs throughout the olfactory system during searchers have speculated and proposed possible func-
development and later decreases to very low levels in tions of markers such as p75NTR and NPY in OECs, un-
the adult rat (Barnett et al., 1993; Franceschini & Bar- equivocal direct evidence that support these assertions
nett, 1996; Gong et al., 1994; Miragall & Dermietzel, is still lacking. These uncertainties underscore the need
1992; Miragall et al., 1988; Miragall et al., 1989; Turner for a greater understanding of OEC biology, before we
& Pérez-Polo, 1992; Vickland et al., 1991). Interestingly, can establish a robust definition for OECs.
p75NTR has been reported to be entirely absent from Despite these uncertainties, all OECs are ultrastruc-
the innermost area of the ONL (Au et al., 2002; Gong turally similar and are derived during ontogeny from
et al., 1994; Treloar et al., 1999; Ubink & Hökfelt, 2000; precursors originating from the olfactory epithelium
Vickland et al., 1991). However, some confusion exists; (Chuah & Au, 1991; Doucette, 1989; Valverde et al.,
for example, OECs were attributed to strong E-NCAM 1992). As the primary olfactory pathway differentiates,
staining in the outer ONL of adult rats (Franceschini & OECs become distributed along the olfactory nerves in
Barnett, 1996), although immunoelectron microscopy the lamina propria as well as within the ONL of the ol-
has shown that, at least in mice, E-NCAM is expressed factory bulb. Olfactory ensheathing cells can be readily
by both OECs and axons during development and is distinguished from other glia by their electron dense cy-
later absent in OECs (Miragall & Dermietzel, 1992; Mi- toplasm, scattered intermediate filaments and irregular
ragall et al., 1988). Similarly, strong p75NTR staining was nuclei (Chuah & Au, 1993; Cuschieri & Bannister, 1975;
reported to be present in OECs in the outer ONL of adult Doucette, 1984; Doucette, 1989; Valverde et al., 1992).
mice (Au et al., 2002), although the localisation of these They ensheath olfactory axons, form the glial limitans
molecules on glial or axonal membranes was not con- of the olfactory bulb and are present throughout the
clusively demonstrated. Furthermore, O4, which was full depth of the ONL (Au et al., 2002; Doucette, 1984;
reported to be co-expressed with p75NTR (Barnett et al., Doucette, 1989; Valverde et al., 1992). They are clearly
1993), was shown by others to be present throughout distinct from interfascicular ACs, which have an elec-
the ONL and in the glomerular layer (Franceschini & tron lucent cytoplasm, multiple bundled intermediate
Barnett, 1996). In fact, recent work has revealed that filaments and are never observed to ensheath axons
OECs do not express O4, but are immunopositive in (Doucette, 1984, 1993).
68 V I N C E N T, W E S T and C H U A H

Antigenic characteristics of OECs in vitro ilar antigenic and morphological phenotypes to those
described previously (Pixley, 1992). These subpopula-
The notion that there are two subpopulations of OECs tions were additionally defined by their E-NCAM ex-
in vivo has been tested on cultured cells based on the as- pression, where SC-like OECs were E-NCAM-negative
sumption that this diversity should be reflected in vitro. and AC-like OECs were E-NCAM-positive (Frances-
Indeed, many studies have demonstrated antigenic het- chini & Barnett, 1996). However, the AC-like subpopu-
erogeneity in cultured OECs, yet comparison between lation was defined differently in cultures derived from
these studies is difficult because each research group the ONL and glomerular layers of adult rat olfactory
has developed unique culturing strategies and different bulb (Li et al., 1998). In this case, AC-like cells were
criteria for defining OECs (Table 1). Olfactory ensheath- described as flat and immunopositive for vimentin and
ing cells have been cultured from olfactory mucosa and fibronectin, with some cells also GFAP-positive (Li et al.,
bulb of both neonatal and adult rats and mice of various 1998). It was conceded that this subpopulation was
strains, using a variety of media, substrates, mitogens, most likely to have been fibroblasts which may be as-
time in culture, number of passages and purification sociated with the outer meningeal surface of the ONL
techniques (Alexander et al., 2002; Barber & Lindsay, (Li et al., 1998; Raisman, 2001), although the portion ex-
1982; Chuah & Au, 1993; Pixley, 1992). Although a num- pressing GFAP would probably have been ACs given
ber of studies have indicated that OEC heterogeneity that the culture was not purified and included adult
exists amongst olfactory bulb-derived OECs (Alexan- glomerular tissue.
der et al., 2002; Barnett et al., 1993; Franceschini & Bar- Hence, there are a number of inconsistencies with
nett, 1996) and to some extent amongst peripherally- the interpretation that cultured OECs are comprised
derived OECs (Barber & Lindsay, 1982; Pixley, 1992), of two subpopulations. (i) The identity of the ‘AC-
there is no experimental data to support a clear anti- like’ subpopulation in each of these studies is question-
genic or functional distinction between these two spa- able. This subpopulation undoubtedly contained an el-
tially different sources of OECs (Au & Roskams, 2003; ement of contaminating ACs and fibroblasts (Li et al.,
Jani & Raisman, 2004; Kumar et al., 2005). 1998; Pixley, 1992; Raisman, 2001) and possibly other
Peripherally located OECs have been observed to dis- unidentified cells types (Barber & Lindsay, 1982). (ii)
play varied intensities of GFAP immunostaining both in The E-NCAM-positive subpopulation may also have
vivo and in vitro (Barber & Lindsay, 1982; Pixley, 1992). contained periglomerular ACs because these cells ex-
The majority of GFAP-positive cells from cultures of press E-NCAM in vivo (Franceschini & Barnett, 1996;
neonatal and adult rat olfactory mucosa were diffusely Miragall & Dermietzel, 1992). (iii) In the absence of lo-
immunostained and had spindly morphology, while a calisation on an ultrastructural level or confirmation
smaller number had intense filamentous staining and by additional techniques, the expression of putative
a flat morphology (Barber & Lindsay, 1982). In another marker molecules based on light microscopy could be
study, OECs cultured from neonatal rat olfactory mu- an artifact in some instances. For example, it has been
cosa were similarly described and were further classi- discovered recently that positive immunoreactivity for
fied based on their resemblance to SCs or to ACs (Pix- O4 on OECs is due to the adhesion of O4-positive ax-
ley, 1992). Schwann cell-like OECs were described as onal membrane to OECs (Wewetzer et al., 2005). Hence,
p75NTR -, S100β- and GFAP-positive (the latter generally the categorisation of OECs into two subpopulations,
weak) with primarily process-bearing morphologies, based solely on immunohistochemical staining, needs
whereas AC-like OECs lacked p75NTR but were GFAP- to be interpreted with caution. (iv) Some of the puta-
positive (mostly fibrous or bright) and gained S100β in tive marker molecules used to define subpopulations
culture, with various flattened morphologies (Pixley, of OECs may not be a consistent feature but demon-
1992). The AC-like OECs comprised only a small per- strates variability depending on OEC interaction with
centage of the total GFAP-positive population (Barber other tissues. For example, the expression of p75NTR
& Lindsay, 1982; Pixley, 1992) and were nearly elimi- by OECs is regulated by axonal contact (Ramón-Cueto
nated from the cultures when care was taken not to har- et al., 1993; Vickland et al., 1991; Wewetzer et al., 2005).
vest central tissue (Pixley, 1992), suggesting that some In the adult ONL, OECs down-regulate expression of
of these AC-like OECs could in fact have been bona fide p75NTR on parts of their membranes which are in direct
ACs. contact with olfactory axons while parts which face the
The categorisation of OECs into SC-like and AC-like pia mater continue to express p75NTR (Vickland et al.,
subpopulations was similarly adopted by others work- 1991). This regulation has also been demonstrated in
ing on OECs cultured from olfactory bulbs (Frances- vitro by two methods. In coculture with olfactory neu-
chini & Barnett, 1996; Li et al., 1998). Olfactory ensheath- rons, p75NTR -positive OECs ensheathed neurites and
ing cells derived from neonatal rat olfactory bulbs lost p75NTR on their membranes where they were in di-
by fluorescence-activated cell sorting of O4-positive rect contact with the neurites (Ramón-Cueto et al., 1993).
cells (Alexander et al., 2002; Barnett et al., 1993) were In freshly dissociated culture, most OECs were p75NTR -
shown to contain subpopulations that displayed sim- negative and had fragments of axonal membrane
Table 1. Strategies of purification and criteria for defining OECs in vitro prior to transplantation into injured CNS tissue.

Research group Animal Tissue source Purification strategy Criteria or description Degree of purity

Ramón-Cueto et al. (1998, 2000), adult Wistar rat OB ; ONL + GL p75+ immunopurification p75+, p75/S100+ ∼ 98%
Ramón-Cueto and Nieto-
Sampedro (1994), Gudiño-
Cabrera and Nieto-Sampedro
(1996), Pascual et al. (2002),
Verdú et al. (1999),
Barnett et al. (2000) adult human OB p75+ immunopurification p75+ high‡
Polentes et al. (2004) adult Sprague-Dawley rat OB Thy1.1/IB4–immunopurification p75+ 75 ± 12%
Gomez et al. (2003) adult Wistar rat ONL + GL Thy1.1/IB4–immunopurification p75/GFAP+ ∼ 90%
Riddell et al. (2004) postnatal§ Fischer rat OB O4+/GalC–immunopurification p75+ highly variable
Smith et al. (2001) postnatal Fischer rat OB O4+/GalC–immunopurification p75+ >95% , predominantly p75+
Ramer et al. (2004) postnatal mouse OM Thy1.1–immunopurification p75/S100/GFAP+ >98%
(GFP transgenic)
Chuah et al. (2003) postnatal Wistar rat ONL + LP mitotic inhibitor p75/GFAP+ 92%
Smith et al. (2002) adult dog ONL shaking, serial passage p75/GFAP/O4+ 92%
Nash et al. (2002) adult Sprague-Dawley rat ON + ONL + GL differential adhesion p75/GFAP+ ∼ 93%
Lakatos et al. (2003) postnatal Fischer rat rostral ONL mitotic inhibitor, shaking, p75+ p75/GFAP+ purified 94% p75/GFAP+,
immunopurification OR unpurified ∼ 75% p75/GFAP+
unpurified (except shaking)
Li et al. (1998, 2003a) adult inbred AS rat ONL + GL none p75/S100+ OECs, ∼ 50% of each
vimentin/fibronectin+ ONFs
Kato et al. (2000) adult human, ON none GFAP/O4/vimentin+, >90% GFAP/vimentin+,
adult Wistar rat S100/GalC/A2B5– ∼ 80% O4+
Radtke et al. (2003) adult pig (HT transgenic) ONL + GL none p75/GFAP/S100+ ∼ 98% p75+
Morphological and functional plasticity of olfactory ensheathing cells

Smale et al. (1996); Boyd et al. (2004) embryonic Wistar rat ONL none S100/p75+ most S100+, part p75+
Pérez-Bouza et al. (1998) adult Lewis rat ON none n.d.# n.d.
Taylor et al. (2001) n.s. n.s. n.s. n.s. n.s.
Boruch et al. (2001) postnatal Sprague-Dawley rat OB n/a-clonal cell line nOEC¶ p75+ 100%
Imaizumi et al. (1998) postnatal Wistar rat ONL n/a-acute dissociated cell n/a n/a
transplantation
Lu et al. (2002) adult Sprague-Dawley rat LP n/a-acute tissue graft n/a n/a
Guntinas-Lichius et al. (2002) adult inbred Lewis rat OM n/a-acute tissue graft n/a n/a

∗ Abbreviations: OB: olfactory bulb, ONL: olfactory nerve layer, GL: glomerular layer, OM: olfactory mucosa, LP: lamina propria, ON: olfactory nerves, GFP: green fluorescent protein,
HT: H-transferase, OECs: olfactory ensheathing cells, ONFs: olfactory nerve fibroblasts, n.s.: not stated, n/a: not applicable, n.d.: not determined.
§up to 7 days after birth; ‡ p75 was rapidly lost in culture and transplanted cell suspensions contained 30% or 45% p75+ cells; ¶ Goodman et al., 1993; #not determined prior to transplantation
but most cells p75/nestin/vimentin+ at 6h post-transplantation.
69
70 V I N C E N T, W E S T and C H U A H

adhering to their surface (Wewetzer et al., 2005). These phatidic acid which induce OECs to adopt a flat mor-
cells became p75NTR -positive after several days when phology probably via downstream assembly of actin
the axon fragments had been phagocytosed (Wewetzer stress fibres and associated focal adhesions (Safavi-
et al., 2005), while the small population of OECs that Abbasi et al., 2001; Suidan et al., 1997). Inactivation of
did not have adherent axon fragments were p75NTR - RhoA by dBcAMP leads to the disassembly of these cy-
positive from the outset (Wewetzer et al., 2005). (v) toskeletal structures causing rapid cytoplasmic retrac-
Finally, the identity of the OEC subpopulations de- tion (Goldman & Abramson, 1990; Ramakers & Moole-
scribed in vitro does not match the description of those naar, 1998; Suidan et al., 1997) and the formation of
in vivo. Cultured OECs are defined by expression of a stellate morphology in OECs. The intermediate fil-
either p75NTR or E-NCAM, whereas OECs in vivo are ament protein GFAP appears to be unaffected by this
thought to express both p75NTR and E-NCAM or other- morphological change in OECs, which does not require
wise to express NPY alone. It would be interesting to in- de novo synthesis or degradation of cytoskeletal proteins
vestigate whether cultured OECs express both p75NTR (Safavi-Abbasi et al., 2001; Vincent et al., 2003). More
and NPY, or whether a NPY-positive/p75NTR -negative recent studies have captured the highly dynamic na-
subpopulation is present and persistent under various ture of OEC morphology in culture using time-lapse
culture conditions. microscopy; individual OECs were observed to switch
Franceschini and Barnett (1996) reported that the from one morphology and back again within an hour
phenotypes displayed by the two putative subpopu- (Van Den Pol & Santarelli, 2003).
lations of OECs were also highly dependent on the From an assessment of all the data now available re-
culture conditions. For example, the expression of garding OEC biology, it would appear that the differ-
E-NCAM and O4 was lost after 7 days in serum- ential expression of markers by OECs, both in vivo and
containing medium and the subpopulations were con- in vitro, is more likely a reflection of their functional
sequently defined in serum-free conditions in which plasticity rather than their existence as discrete, inex-
they were more distinguishable. However, it should changable subpopulations. The stongest data in sup-
be noted that E-NCAM and O4 may be attributed to port of this interpretation are the observations that (i)
fragments of axonal membrane and their “disappear- ultrastructually uniform OECs reside throughout the
ance’’ may be a consequence of phagocytosis by OECs full depth of the ONL (Au et al., 2002; Valverde et al.,
(Wewetzer et al., 2005), a process which would have 1992; Doucette, 1989), (ii) in vitro studies have demon-
progressed more slowly in serum-free conditions (Mor- strated that expression of widely used OEC markers,
ris et al., 2003). Cells adopted divergent morpholo- e.g. p75NTR , can be regulated by OEC interaction with
gies when long-term cultures were switched between extrinsic factors such as axonal contact (Wewetzer et al.,
different types of media (Alexander et al., 2002). Un- 2005; Ramón-Cueto et al., 1993; Vickland et al., 1991),
der these conditions E-NCAM expression oscillated and (iii) OECs display morphological plasticity in vitro
markedly but p75NTR and GFAP remained constant (Vincent et al., 2003; Van Den Pol & Santarelli, 2003) and
(Alexander et al., 2002). Increasing cell density also following transplantation into injured CNS tissue (to be
correlated with increasing expression of p75NTR and discussed below).
GFAP in serum-containing medium, although p75NTR
decreased at later passages while E-NCAM increased
Relationship between olfactory ensheathing cells
(Sonigra et al., 1999).
and developing olfactory nerves
These findings suggest that OECs have a malleable
phenotype that is dependent on environmental stimuli The axon pathfinding of olfactory neurons involves a
(Au & Roskams, 2003; Chuah & West, 2002; Doucette, range of guidance mechanisms which are still the sub-
1995). A recent study by our laboratory showed that ject of intense research (reviewed by Key & St John,
OECs derived from the olfactory mucosa and ONL 2002). In mammals, the direct role of OECs in this pro-
underwent rapid and reversible change between flat cess is yet to be clearly defined, although glia cells that
morphology in the presence of serum and process- play a key role in sorting olfactory axons are present in
bearing morphology in the absence of serum (Vin- invertebrates such as the moth, Manduca sexta (Rossler
cent et al., 2003). Purified OECs were 87–90% flat in et al., 1999). Olfactory axon growth in mammals takes
serum-containing medium and became 78–84% bipo- place in a series of steps, regulated by distinct guid-
lar or multipolar in serum-free medium within 24 h of ance cues some of which are provided by OECs. First,
the change in conditions. Rapid morphological change axogenesis occurs and new axons emerge from the ol-
in OECs was mediated by the small guanosine triphos- factory epithelium into the lamina propria. Axons from
phatase RhoA signalling cascade which induces equiv- the same neuroepithelial zone fasciculate into olfactory
alent changes in AC morphology and is important in nerve bundles, which are enveloped by cytoplasmic
cytoskeletal organisation in a variety of other cells (Hall, processes of OECs along their trajectory in the lamina
1998; Ridley & Hall, 1992; Vincent et al., 2003; Yan et al., propria. As the axons approach the bulb, they segregate
2003). RhoA is activated by endothelin-1 and lysophos- broadly into dorsally and ventrally projecting nerve
Morphological and functional plasticity of olfactory ensheathing cells 71

bundles. Once within the ONL, the olfactory nerves ulation expressing lactosamine-containing glycans con-
defasciculate and sort into smaller bundles. It has been verges on this region which has high expression of all
shown that axons expressing the same odorant receptor three molecules (Crandall et al., 2000; Schwarting et al.,
gene converge and target to specific glomeruli which 2000). Olfactory ensheathing cells also differentially ex-
are topographically fixed (Belluscio et al., 2002; Bozza press galectin-7 (Storan et al., 2004) and p75NTR (Cran-
et al., 2002; Mombaerts et al., 1996). The elongation of dall et al., 2000) during development, although roles
olfactory axons in the lamina propria may be attributed for these putative guidance molecules have not been
largely to the behaviour of OECs (Key & St John, 2002), established.
whereas OEC involvement in the targeting of axon bun- The role of OECs in guiding olfactory axonal growth
dles in the ONL to specific glomeruli is unlikely. may be attributed also to their expression of glia-
The trajectory of olfactory nerves established during derived nexin, a protease inhibitor which also possesses
development are formed by conduits of OEC processes axonal-promoting properties (Reinhard et al., 1988). In-
expressing cell adhesion and extracellular molecules terestingly, the ONL of the olfactory bulb is the only
such as galectin-1, β2-laminin, NCAM, E-NCAM, L1 site in the mammalian CNS where ACs also express
and collagen IV (Crandall et al., 2000; Miragall et al., glia-derived nexin (Scotti et al., 1994). Whether this re-
1989; Whitesides & LaMantia, 1996). These growth- flects some special property of olfactory bulb ACs as
permissive tracks laid down by OECs are thought to fa- a result of their close spatial association with OECs, or
cilitate large-scale fasciculation and elongation of new whether OEC function is influenced by surrounding
axons towards their central targets (Chuah & Au, 1993; ACs remains to be elucidated.
Key & St John, 2002; Whitesides & LaMantia, 1996). It can be summarised from these studies that OECs
However, axon elongation is optimised in conditions express a complex repertoire of molecules that are
which limit adhesion (St John et al., 2000), such as known to be developmentally significant and assume
by the expression of polysialic acid (PSA) on NCAM functionally crucial roles both while they are a com-
(E-NCAM) and EphA5 (St John et al., 2000). These ponent of the peripheral olfactory nerves as well as a
molecules are expressed during the developmental pe- cellular constituent of the ONL in the bulb. In light of
riod corresponding to massive axon growth and are their expression of axon-promoting molecules and the
postnatally downregulated (Miragall et al., 1989; St John general guidance role that they play in the development
et al., 2000). EphA5 is expressed by olfactory neurons of the primary olfactory pathway, OECs have become
and OECs and is essential for olfactory axon elongation an obvious experimental tool in the repair of the injured
in vitro (St John et al., 2000). EphA5 belongs to a family CNS. The following sections will discuss structural in-
of repulsive factors but is thought in this case to facili- teraction between OECs and other cell types in such a
tate direct and unbranching axon growth by regulating novel environment.
adhesion strength (St John et al., 2000).
In addition to acting as a suitable substratum, OECs
Olfactory ensheathing cells in injured central
are thought to migrate in tandem with the growing ol-
nervous tissue
factory axons. The guidance mechanisms controlling
OEC migration to the presumptive olfactory bulb dur- Several studies have indicated that OEC transplanta-
ing development are ultimately responsible for the di- tion holds promise as a potential therapy for spinal
rectional guidance of the pioneer axons and subsequent cord injury, although other studies have suggested
establishment of the olfactory nerve tracts (Key & St that OECs may not always be beneficial. Compari-
John, 2002). This migration may be mediated by a bulb- son between studies is made difficult by a multitude
derived chemoattractant (Liu et al., 1995), similar to that of approaches by the different research groups, for
required by olfactory axons in the moth, Manduca sexta example the various purification strategies and cri-
(Oland et al., 2003). The same or related factors may be teria used for defining OECs (Table 1). In the most
responsible in rats for attracting neuronal precursors successful experiments, adult rats that received OEC
from the subventricular zone to their olfactory destina- transplants recovered climbing ability and sensori-
tions (Liu & Rao, 2003). motor reflexes after complete spinal cord transection
At the distal end of olfactory nerves, OECs create an (Ramón-Cueto et al., 2000), respiratory function after
axon-sorting domain in the ventral ONL where olfac- cervical hemisection (Li et al., 2003a; Polentes et al.,
tory axons initially cross the PNS-CNS boundary (Cran- 2004) and bladder control after lumbosacral dorsal root
dall et al., 2000). Olfactory ensheathing cells differen- transection (Pascual et al., 2002). This functional re-
tially express galectin-1 and its ligand β2-laminin and covery correlated with anatomical and activity-based
the chemorepulsive factor Sema3A in the ONL (Cran- regeneration in the associated axon tracts, including
dall et al., 2000; Schwarting et al., 2000; Pasterkamp et al., long distance regrowth of some axons (Pascual et al.,
1998). The subpopulation of olfactory axons express- 2002; Polentes et al., 2004; Ramón-Cueto et al., 2000;
ing the Sema3A receptor, neuropilin-1, is repelled from Ramón-Cueto & Nieto-Sampedro, 1994; Ramón-Cueto
the ventral Sema3A-positive region, while the subpop- et al., 1998). Furthermore, OECs transplanted from
72 V I N C E N T, W E S T and C H U A H

rats, dogs, pigs and humans into demyelinating le- section (Lee et al., 2004). In studies where axonal re-
sions in the rat spinal cord promoted remyelination generation was successful, OECs adopted a spindle-
of axons and restored impulse conduction (Barnett shaped morphology forming a bridge or scaffold, facil-
et al., 2000; Imaizumi et al., 2000a; Imaizumi et al., itating the growth of axons across the glial scar (Boruch
2000b; Imaizumi et al., 1998; Kato et al., 2000; Li et al., et al., 2001; Li et al., 1998; Pérez-Bouza et al., 1998). How-
1998; Smith et al., 2002). Similar results were obtained ever, the efficacy of OECs is not consistently guaranteed
when OECs were transplanted from pig into non- (Gudiño-Cabrera et al., 2000; Takami et al., 2002) and is
human primate demyelinating lesions (Radtke et al., highly contentious at the dorsal root entry zone, be-
2004). ing supported by many studies (Li et al., 2004; Navarro
Although OECs are able to promote regeneration in et al., 1999; Pascual et al., 2002; Ramón-Cueto & Nieto-
the CNS (Guntinas-Lichius et al., 2002; Li et al., 2003b; Sampedro, 1994; Taylor et al., 2001) but disputed by
Verdú et al., 1999), the precise mechanisms mediating others (Gomez et al., 2003; Ramer et al., 2004a; Riddell
this process remain ill-defined. Using an in vitro model et al., 2004). These discrepancies warrant further inves-
of axonal injury, it has been shown that OECs formed a tigation into the cellular mechanisms of OEC-mediated
physical substrate for the growth of post-injury neurite repair. It is possible that the degree of inflammation
sprouts (Chung et al., 2004). Neurite sprouting was most present in the injury site coupled with the manner of
exuberant when OECs were allowed to contact directly interaction between OECs and astrocytes influences the
the injured neurons. If OECs and injured neurons were extent of repair. Some researchers have argued that
physically separated but allowed to share the same cul- transplantation of OECs by means of cell injections
ture medium, neurite sprouting was reduced but not (Ramer et al., 2004a; Riddell et al., 2004) causes exten-
blocked completely. Hence, the results suggested that sive damage of spinal cord tissue which impedes axonal
both secretion of soluble factors and direct membrane repair and interferes with optimal interaction between
interaction with injured axons are likely to be involved OECs and ACs (Li et al., 2004). Instead, the application
in OEC-induced axon regeneration (Chung et al., 2004). of OECs within an endogenous matrix to the cut sur-
In a coculture assay for neurite growth of retinal gan- face of a dorsal root and subsequent stabilisation with
glion cells, p75NTR -immunoselected OECs were found fibrin glue afforded the optimal condition for repair (Li
to be more efficacious in promoting neurite outgrowth et al., 2003a; Li et al., 2004).
than p75NTR -negative cells from the same tissue or Recent in vitro studies have shown that there is a
than a mixed population (Kumar et al., 2005). Whether marked difference in the way that OECs and SCs in-
p75NTR -positive cells were harvested from the ONL or teract with ACs (Lakatos et al., 2000; Van Den Pol
from peripherally located olfactory nerve rootlets made & Santarelli, 2003). Olfactory ensheathing cells were
no difference to neurite outgrowth in this assay (Kumar shown to migrate and intersperse with ACs, whereas
et al., 2005). The precise function of p75NTR expressed SCs and ACs remained segregated (Lakatos et al., 2000).
by OECs in promoting neurite growth remains to be Recent evidence suggests that differential regulation of
elucidated. N-cadherin in OECs and SCs may be the reason for
In in vivo situations, where OECs necessarily interact their differing interaction with ACs (Fairless et al., 2005).
with a greater variety of cell types and within a three The affinity which OECs show towards ACs could un-
dimensional matrix, additional factors come into play. derly their ability to induce changes in host reactive
It appears that restricted migration and optimal phys- ACs at the injury site. It has been shown that trans-
ical orientation of OECs at the injury site may play a planted OECs are capable of migrating both longitudi-
role in inducing a favourable environment for regener- nally and laterally within the injured CNS and can in-
ation. (Li et al., 1998). Interestingly, studies that involve termingle with ACs within the glial scar (Ramón-Cueto
transplantation of OECs into CNS regions other than et al., 1998). Furthermore, their ability to migrate within
the spinal cord, have reported more widespread mi- the injured spinal cord is greater than that of SCs (Li
gration of OECs (Gudiño-Cabrera & Nieto-Sampedro, et al., 1998). However, it should be noted that migra-
1996; Gudiño-Cabrera et al., 2000). For example, when tion of OECs appears to be limited by the extent of the
OECs were transplanted to the hippocampal CA1 re- glial scar. In vivo tracking of OECs labelled with super-
gion, they could be found one month later in distant paramagnetic iron oxide nanoparticles using magnetic
loci such as the laterodorsal thalamic nuclei, internal resonance imaging showed that OECs were not able
capsule and arcuate nucleus (Gudiño-Cabrera & Nieto- to cross the glial scar present at the site of complete
Sampedro, 1996). This migratory behaviour was shown spinal transection (Lee et al., 2004). Nevertheless, there
to be detrimental to regeneration when OECs were im- is general agreement that compared to those which
planted into the brainstem and they migrated in a di- received SCs, injured spinal cords which were trans-
rection opposite to the intended axonal target (Gudiño- planted with OECs had comparatively less astrogliosis
Cabrera et al., 2000). as measured by hypertrophy and expression of chon-
Migration of OECs appears to be more restricted in droitin sulfate proteoglycan and GFAP (Garcia-Alias
the spinal cord, particularly in cases of complete tran- et al., 2004; Lakatos et al., 2003).
Morphological and functional plasticity of olfactory ensheathing cells 73

Fig. 2. Scanning electron micrograph of underside of porous inserts facing astrocytes (A). The pores measure 1 µm in diameter
and two of the pores demonstrate the presence of OEC processes emerging from the other side. In the rare instance, entire OECs
had squeezed themselves through the pores and are apparently hanging on by their narrow processes (B). Scale bar = 1 µm
(A) and 4 µm (B).

In a study involving lesion to the dorsal root en- forming growth factor β (O’Toole et al., 2005). The
try zone in which there was successful regeneration of porous inserts allowed the two cell types to share the
about 10% of axotomised fibres, it was shown that trans- same medium in the absence of direct contact. Follow-
planted OECs interacted with host ACs and SCs of the ing 24 hours’ culture, processes of some OECs could be
dorsal root to form a ladder-like structure that allowed observed traversing the 1 µm diameter pores and ex-
ingrowth of nerve fibres across the normally inhibitory tending towards the ACs (Fig. 2A). In rare instances
PNS-CNS interface (Li et al., 2004). Using an in vitro entire OECs were able to squeeze through the 1 µm di-
model which allows populations of cells to be physi- ameter pores and to hang on to the underside of the
cally separated but sharing the same culture medium inserts by their processes (Fig. 2B).
(Chung et al., 2004), we have obtained some preliminary In summary, transplanted OECs display morpholog-
results that suggest that interaction between OECs and ical plasticity in response to as yet unidentified cues
reactive astrocytes may be mediated by soluble factors. from the novel injured CNS environment. It is likely that
OECs were cultured from neonatal rat ONL and ol- the morphological plasticity demonstrated by OECs re-
factory mucosa and purified as previously described. flects an equally robust functional plasticity in their in-
(Vincent et al., 2003), and with approval from the Ani- teraction with CNS tissues.
mal Experimentation Ethics Committee of the Univer-
sity of Tasmania. Olfactory ensheathing cells were cul-
tured overnight in porous Falcon Cell Culture Inserts Other possible functional roles of olfactory
(catalog # 353104, Becton Dickinson Labware, Franklin ensheathing cells
Lakes, NJ) overlying ACs, which were pre-activated It is well accepted that olfactory neurons lining the
by scratch wounding and exposure to 10 ng/mL trans- nasal cavity are continuously renewed throughout an
74 V I N C E N T, W E S T and C H U A H

animal’s life (Graziadei & Graziadei, 1979); some of vent the turbidity induced in the culture medium when
them have a lifespan of about one month while oth- the latter is infected with attenuated Staphylococcus au-
ers are known to survive for much longer (Mackay-Sim reus and Escherichia coli (JM109 strain) (Vincent et al.,
& Kittel, 1990). As mature olfactory neurons die, they 2005). Further investigation was done to test whether
are replaced by a new population of cells derived from OECs were capable of phagocytosing whole bacteria
globose basal cells residing in the olfactory epithelium in vitro. Fluorescently-labelled Microccoccus luteus (M.
(Mackay-Sim & Kittel, 1991). In the unperturbed ani- luteus; dead, Gram-positive; EnzCheck Lysozyme As-
mal, it has been observed by electron microscopy that say kit, Molecular Probes, Eugene, OR) were incubated
small numbers of macrophages are present in the olfac- with OECs for 24 h and the living cultures were viewed
tory epithelium. (Graziadei & Graziadei, 1979). These under fluorescence microscopy. M. luteus were associ-
macrophages were assumed to be responsible for dis- ated preferentially with the cell bodies of many OECs
posing of degenerating epithelial cells under normal in each culture, particularly surrounding the nucleus
physiological conditions. However, they are not the (Fig. 3A and B). Interestingly, internalised M. luteus ap-
only cell type that is capable of phagocytosing cel- peared to have been protected from cross-reaction with
lular debris in the olfactory system. Bulbectomy and
infection by neurotropic pathogens induce rapid and
massive death of olfactory neurons. Removal of the re-
sulting cell debris and pathogens in the acute phase is
marked by an increase in the number of macrophages
infiltrating the degenerating epithelium. (Suzuki et al.,
1995). In addition, supporting cells of the olfactory ep-
ithelium also become phagocytic as indicated by the
presence of phagosomes and apoptotic bodies in the
cytoplasm (Suzuki et al., 1996). Not only are they ac-
tive during the acute phase of degeneration, phagocytic
supporting cells can be observed up to two months after
bulbectomy (Suzuki et al., 1996).
Whilst macrophages and supporting cells are in-
volved in clearance of dead cell bodies in the epithe-
lium, OECs appear to contribute to phagocytosis of ax-
onal debris in the lamina propria and olfactory bulb
(Chuah et al., 1995). Electron microscopy showed that
following zinc sulphate irrigation of the nasal cav-
ity which destroyed much of the olfactory epithelium,
some OECs forming the glia limitans of the olfactory
bulb were observed to contain phagosomes and cellu-
lar debris in their cytoplasm (Chuah et al., 1995). The
phagocytic activity of bulb-derived OECs has been re-
cently confirmed in an in vitro study which showed
that the O4 immunoreactivity of OECs was actually
due to O4-positive axonal fragments adhering to OECs
(Wewetzer et al., 2005). Using a novel two-step label-
ing protocol based on antibody internalisation, Wewet-
zer and co-workers showed that the O4-positive ax-
onal fragments were phagocytosed by OECs, and as
immunoreactivity for O4 decreased, the OECs upregu-
lated their expression of p75NTR .
Based on these findings, it is logical to ask if OECs
are capable of showing anti-bacterial activity, such as
when infected olfactory axons degenerate and release Fig. 3. Green fluorescently labeled M.luteus surround the nu-
pathogens, thus acting as a member of the innate im- cleus of OECs stained by Hoescht Blue dye (A); cytoplasm
mune system. Given that OECs are present in the lam- of the OECs stain positive for S100 (B). In (C) immunostain-
ina propria beneath the olfactory epithelium, it would ing for Lyz show different intensities of labeling in two OECs.
be reasonable to assume that they probably form a The M.luteus adhering on the external surface of the unperme-
line of defence secondary to olfactory supporting cells abilised OECs or to the coverslip show cross-reactivity with
and macrophages. In preliminary experiments from our anti-Lyz (bright orange) while the internalized M.luteus are
laboratory, it was found that OECs were able to pre- fluorescently green. Scale bar = 10 µm.
Morphological and functional plasticity of olfactory ensheathing cells 75

anti-lysozyme (Lyz) antibodies in the unpermeabilised reported that in the aftermath of bulbectomy and fol-
cells (Fig. 3C), in contrast to M. luteus found attached lowing infiltration of IL-6 producing macrophages into
to the external surface of OECs or to the coverslip. the olfactory lamina propria, OECs were shown to up-
The finding that OECs could have antibacterial ac- regulate their expression of IL-6 receptors (Nan et al.,
tivity in culture is intriguing given that the olfactory 2001). These authors proposed that IL-6 may be in-
system is the only neuroepithelium in the body that is volved in initiating intracellular signalling processes
directly exposed to the external environment. Mecha- that ultimately lead to OECs establishing extracellu-
nisms which are known to protect this tissue from in- lar matrices which facilitate olfactory axonal regrowth
fection include immunogenic mucosal secretion, and (Nan et al., 2001). Such a capability could be function-
lactoferrin and Lyz secretion from Bowman’s glands ally significant as well, when OECs are transplanted
(Getchell & Getchell, 1991). In addition, recent studies into an acute injury site in the CNS.
from our laboratory have shown that OECs produce Another protein, which could prove to be function-
mRNA for Lyz and are immunopositive for Lyz protein ally significant in the context of OECs as a form of cell
both in vivo and in vitro (Vincent et al., 2005). Although therapy in CNS injury, is connective tissue growth fac-
it was not stated by the authors, a previous immuno- tor (Ctgf). In the unperturbed olfactory system, Ctgf has
histochemical study in the rat and salamander showed been observed in the embryonic mouse olfactory ep-
that OECs appeared to be immunopositive for Lyz (see ithelium (Surveyor & Brigstock, 1999). Ctgf is involved
Fig. 6h in Getchell & Getchell, 1991). A role for Lyz in diverse processes in the nervous system which in-
activity in OECs is consistent with the susceptibility clude angiogenesis, fibrotic and glial scarring, and ex-
of the primary olfactory pathway to external chemi- tracellular matrix remodelling in normal and injured
cal and pathogenic insults. A recent study showed that tissue. (Hertel et al., 2000; Schwab et al., 2001). In a re-
pneumococcal infection can occur from the olfactory cent study, it was reported that the region of the dorsal
epithelium to the olfactory bulb via transport along ol- column in which OECs were injected had deposits of
factory nerves, but that this route is limited by an un- laminin and showed the presence of numerous new
known mechanism (van Ginkel et al., 2003). If OECs blood vessels. (Ramer et al., 2004b). It is tempting to
can be proved to contribute to a neuroprotective role in speculate that proteins expressed by OECs, such as Ctgf
the olfactory system, in the future it may be possible to and vascular endothelial growth factor (Au & Roskams,
prime or activate them by intranasal delivery of specific 2003), may be involved in the reported neoangiogene-
drugs or genes (Frey, 2002; Thorne et al., 2004) in order to sis. Intriguingly, although neovascularisation is gener-
protect people at high risk of contracting meningitis or ally thought to be beneficial to repair, in this particu-
encephalitis. lar study, axotomised dorsal roots failed to regenerate
In a recent microarray study, factors involved in the (Ramer et al., 2004b).
immune response and inflammation have been de- An immune function in OECs may have significant
tected as transcripts enriched in OECs, including the implications for the transplantation of these cells as a
chemokines Cxcl1 (also known as Gro1) and Ccl2 (also therapy for CNS injury. Physical trauma of the CNS
known as MCP1). Ccl2 is a monocyte chemoattractant is often accompanied by a massive inflammatory re-
and Cxcl1 is a neutrophil chemoattractant that is also in- sponse which is thought to cause secondary damage
volved in glial development (Dong & Benveniste, 2001). leading to neuron loss (Bareyre & Schwab, 2003; Blight,
The protein levels for these factors are low or absent in 1992; Carmel et al., 2001). The anti-inflammatory drug
OECs in vitro and in normal olfactory tissue (Vincent methylprednisolone is used in the treatment of hu-
et al., 2005), but this is consistent with the expression man spinal cord injury in an attempt to manage this
of Ccl2 in Schwann cells, which is normally low in the secondary damage. Concievably, transplanted OECs
sciatic nerve and is upregulated in response to injury may promote regeneration via a similar mechanism,
(Toews et al., 1998). Astrocytes in the spinal cord pro- by modulating neuroinflammation in the injured CNS.
duce Cxcl1 in a spatiotemporal manner to help direct Indeed, OECs used in a combination treatment with
oligodendrocyte precursor maturation and migration methylprednisolone in lesioned rat spinal cord were
(Tran & Miller, 2003). Although immunohistochemical found to have a synergistic effect on axonal regenera-
staining showed that OECs do not appear to produce tion and functional recovery (Nash et al., 2002).
detectable amounts of Ccl2 or Cxcl1 proteins in the nor- In summary, increasing understanding of the func-
mal adult olfactory system, a role for these factors may tional plasticity of OECs may well reveal that this
exist when these cells are transplanted into the injury unique glial cell could have multiple functions extend-
site in the CNS. ing beyond its role in the normal olfactory system.
Furthermore, it is likely that OECs are themselves There is now much experimental evidence that shows
capable of responding to immune factors, as suggested the regenerative property of the olfactory system can
by their high level of expression for the interleukin 6 replace the inhibitory nature of the injured CNS by
(IL-6)-responsive transcription factor, Cebpb (Vincent OEC transplantation. Whilst their use in promoting re-
et al., 2005). This notion is supported by a study which pair in experimentally induced CNS injury reflects their
76 V I N C E N T, W E S T and C H U A H

versatility, their future development into a reliable clin- BOYD, J. G., LEE, J., SKIHAR, V., DOUCETTE, R. &
ical therapy can only be assured by a better understand- KAWAJA, M. D. (2004) LacZ-expressing olfactory en-
ing of their cell biology. sheathing cells do not associate with myelinated axons
after implantation into the compressed spinal cord. Pro-
ceedings of the National Academy of Science 101, 2162–2166.
Acknowledgments BOYD, J. G., SKIHAR, V., KAWAJA, M. & DOUCETTE, R.
(2003) Olfactory ensheathing cells: Historical perspective
Research conducted by the authors was supported by and therapeutic potential. Anatomical Record 271B, 49–60.
grants from the National Health and Medical Research BOZZA, T., FEINSTEIN, P., ZHENG, C. & MOMBAERTS,
Council and the International Institute for Research in P. (2002) Odorant receptor expression defines functional
Paraplegia. MIC would like to express her gratitude to units in the mouse olfactory system. Journal of Neuro-
Albert Farbman for his mentoring which started her on science 22, 3033–30343.
CARMEL, J. B., GALANTE, A., SOTEROPOULOS, P.,
a career in basic research.
TOLIAS, P., RECCE, M., YOUNG, W. & HART, R.
P. (2001) Gene expression profiling of acute spinal cord
injury reveals spreading inflammatory signals and neu-
References ron loss. Physiological Genomics 7, 201–213.
ALEXANDER, C. L., FITZGERALD, U. F. & BARNETT, CHUAH, M. I. & AU, C. (1991) Olfactory Schwann cells are
S. C. (2002) Identification of growth factors that promote derived from precursor cells in the olfactory epithelium.
long-term proliferation of olfactory ensheathing cells and Journal of Neuroscience Research 29, 172–180.
modulate their antigenic phenotype. Glia 37, 349–364. CHUAH, M. I. & AU, C. (1993) Cultures of ensheathing
ASTIC, L., PELLIER-MONNIN, V. & GODINOT, F. (1998) cells from neonatal rat olfactory bulbs. Brain Research 601,
Spatio-temporal patterns of ensheathing cell differenti- 213–220.
ation in the rat olfactory system during development. CHUAH, M. I., CHOI-LUNDBERG, D., WESTON, S.,
Neuroscience 84, 295–307. VINCENT, A. J., CHUNG, R. S., VICKERS, J. C. &
AU, E. & ROSKAMS, A. J. (2003) Olfactory ensheathing cells WEST, A. K. (2004) Olfactory ensheathing cells promote
of the lamina propria in vivo and in vitro. Glia 41, 224–236. collateral axonal branching in the injured adult rat spinal
AU, W. W., TRELOAR, H. B. & GREER, C. A. (2002) Sub- cord. Experimental Neurology 185, 15–25.
laminar organization of the mouse olfactory bulb nerve CHUAH, M. I., TENNENT, R. & JACOBS, I. (1995) Re-
layer. Journal of Comparative Neurology 446, 68–80. sponse of olfactory Schwann cells to intranasal zinc
BARBER, P. C. & LINDSAY, R. M. (1982) Schwann cells of sulfate irrigation. Journal of Neuroscience Research 42,
the olfactory nerves contain glial fibrillary acidic protein 470–478.
and resemble astrocytes. Neuroscience 7, 3077–90. CHUAH, M. I. & WEST, A. K. (2002) Cellular and molecu-
BAREYRE, F. M. & SCHWAB, M. E. (2003) Inflammation, lar biology of ensheathing cells. Microscopy Research and
degeneration and regeneration in the injured spinal cord: Technique 58, 216–227.
insights from DNA microarrays. Trends in Neuroscience CHUNG, R. S., WOODHOUSE, A., FUNG, S., DICKSON,
26, 555–563. T. C., WEST, A. K., VICKERS, J. C. & CHUAH, M.
BARNETT, S. C. (2004) Olfactory ensheathing cells: unique I. (2004) Olfactory ensheathing cells promote neurite
glial cell types? Journal of Neurotrauma 21, 375–382. sprouting of injured axons in vitro by direct cellular con-
BARNETT, S. C., ALEXANDER, C. L., IWASHITA, Y., tact and secretion of soluble factors. Cellular and Molecular
GILSON, J. M., CROWTHER, J., CLARK, L., DUNN, Life Sciences 61, 1238–1245.
L. T., PAPANASTASSIOU, V., KENNEDY, P. G. & CRANDALL, J. E., DIBBLE, C., BUTLER, D., PAYS,
FRANKLIN, R. J. (2000) Identification of a human olfac- L., AHMAD, N., KOSTEK, C., PUSCHEL, A. W. &
tory ensheathing cell that can effect transplant-mediated SCHWARTING, G. A. (2000) Patterning of olfactory
remyelination of demyelinated CNS axons. Brain 123, sensory connections is mediated by extracellular matrix
1581–1588. proteins in the nerve layer of the olfactory bulb. Journal
BARNETT, S. C. & CHANG, L. (2004) Olfactory ensheath- of Neurobiology 45, 195–206.
ing cells and CNS repair: going solo or in need of a friend? CUSCHIERI, A. & BANNISTER, L. H. (1975) The devel-
Trends in Neuroscience 27, 54–60. opment of the olfactory mucosa in the mouse: electron
BARNETT, S. C., HUTCHINS, A. M. & NOBLE, M. (1993) microscopy. Journal of Anatomy 119, 471–498.
Purification of olfactory nerve ensheathing cells from the DONATO, R. (1999) Functional roles of S100 proteins,
olfactory bulb. Developmental Biology 155, 337–350. calcium-binding proteins of the EF-hand type. Biochimica
BELLUSCIO, L., LODOVICHI, C., FEINSTEIN, P., et Biophysica Acta 1450, 191–231.
MOMBAERTS, P. & KATZ, L. C. (2002) Odorant recep- DONG, Y. & BENVENISTE, E. N. (2001) Immune function
tors instruct functional circuitry in the mouse olfactory of astrocytes. Glia 36, 180–190.
bulb. Nature 419, 296–300. DOUCETTE, J. R. (1984) The glial cells in the nerve fiber layer
BLIGHT, A. R. (1992) Macrophages and inflammatory dam- of the rat olfactory bulb. Anatomical Record 210, 385–391.
age in spinal cord injury. Journal of Neurotrauma 9, 83–91. DOUCETTE, J. R. & DEVON, R. (1993) Olfactory ensheath-
BORUCH, A. V., CONNERS, J. J., PIPITONE, M., ing cells: factors influencing the phenotype of these glial
DEADWYLER, G., STORER, P. D., DEVRIES, G. H. & cells. In: Biology and Pathology of Astrocyte-Neuron Inter-
JONES, K. J. (2001) Neurotrophic and migratory proper- actions (edited by Sergey Fedoroff, Berhard H.J. Juurlink,
ties of an olfactory ensheathing cell line. Glia 33, 225–229. Ronald Doucette), pp. 117–124. New York: Plenum Press.
Morphological and functional plasticity of olfactory ensheathing cells 77

DOUCETTE, J. R., KIERNAN, J. A. & FLUMERFELT, B. GRAZIADEI, P. P. & GRAZIADEI, G. A. (1979) Neuroge-
A. (1983) The re-innervation of olfactory glomeruli fol- nesis and neuron regeneration in the olfactory system
lowing transection of primary olfactory axons in the cen- of mammals. I. Morphological aspects of differentiation
tral or peripheral nervous system. Journal of Anatomy 137, and structural organization of the olfactory sensory neu-
1–19. rons. Journal of Neurocytology 8, 1–18.
DOUCETTE, R. (1989) Development of the nerve fiber layer GUDI ÑO-CABRERA, G. & NIETO-SAMPEDRO, M. (1996)
in the olfactory bulb of mouse embryos. Journal of Com- Ensheathing cells: Large scale purification from adult ol-
parative Neurology 285, 514–527. factory bulb, freeze-preparation and migration of trans-
DOUCETTE, R. (1990) Glial influences on axonal growth in planted cells in adult brain. Restorative Neurology and Neu-
the primary olfactory system. Glia 3, 433–449. roscience 10, 25–34.
DOUCETTE, R. (1991) PNS-CNS transitional zone of the GUDI ÑO-CABRERA, G., PASTOR, A. M., DE LA
first cranial nerve. Journal of Comparative Neurology 312, CRUZ, R. R., DELGADO-GARCIA, J. M. & NIETO-
451–466. SAMPEDRO, M. (2000) Limits to the capacity of trans-
DOUCETTE, R. (1993) Glial cells in the nerve fiber layer of the plants of olfactory glia to promote axonal regrowth in the
main olfactory bulb of embryonic and adult mammals. CNS. Neuroreport 11, 467–471.
Microscopy Research and Technique 24, 113–130. GUNTINAS-LICHIUS, O., WEWETZER, K., TOMOV,
DOUCETTE, R. (1995) Olfactory ensheathing cells: poten- T. L., AZZOLIN, N., KAZEMI, S., STREPPEL, M.,
tial for glial cell transplantation into areas of CNS injury. NEISS, W. F. & ANGELOV, D. N. (2002) Transplan-
Histology and Histopathology 10, 503–507. tation of olfactory mucosa minimizes axonal branching
FAIRLESS, R., FRAME, M. C. & BARNETT, S. C. (2005) and promotes the recovery of vibrissae motor perfor-
N-cadherin differentially determines Schwann cell and mance after facial nerve repair in rats. Journal of Neu-
olfactory ensheathing cell adhesion and migration re- roscience 22, 7121–7131.
sponses upon contact with astrocytes. Molecular and Cel- HALL, A. (1998) Rho GTPases and the actin cytoskeleton.
lular Neuroscience 28, 253–263. Science 279, 509–514.
FRANCESCHINI, I. A. & BARNETT, S. C. (1996) Low- HERTEL, M., TRETTER, Y., ALZHEIMER, C. &
affinity NGF-receptor and E-N-CAM expression define WERNER, S. (2000) Connective tissue growth fac-
two types of olfactory nerve ensheathing cells that share tor: a novel player in tissue reorganization after brain
a common lineage. Developmental Biology 173, 327–343. injury? European Journal of Neuroscience 12, 376–380.
FREY, W. H., 2ND (2002) Intranasal delivery. Bypassing the IMAIZUMI, T., LANKFORD, K. L., BURTON, W. V.,
blood-brain barrier to deliver therapeutic agents to the FODOR, W. L. & KOCSIS, J. D. (2000a) Xenotrans-
brain and spinal cord. Drug Delivery Technology 2, 46– plantation of transgenic pig olfactory ensheathing cells
49. promotes axonal regeneration in rat spinal cord. Nature
GARCIA-ALIAS, G., LOPEZ-VALES, R., FORES, J., Biotechnology 18, 949–953.
NAVARRO, X. & VERD U , E. (2004) Acute transplanta- IMAIZUMI, T., LANKFORD, K. L. & KOCSIS, J. D.
tion of olfactory ensheathing cells or Schwann cells pro- (2000b) Transplantation of olfactory ensheathing cells
motes recovery after spinal cord injury in the rat. Journal or Schwann cells restores rapid and secure conduction
of Neuroscience Research 75, 632–641. across the transected spinal cord. Brain Research 854, 70–
GETCHELL, M. L. & GETCHELL, T. V. (1991) Immuno- 78.
histochemical localization of components of the immune IMAIZUMI, T., LANKFORD, K. L., WAXMAN, S. G.,
barrier in the olfactory mucosae of salamanders and rats. GREER, C. A. & KOCSIS, J. D. (1998) Transplanted ol-
Anatomical Record 231, 358–374. factory ensheathing cells remyelinate and enhance ax-
GOLDMAN, J. E. & ABRAMSON, B. (1990) Cyclic AMP- onal conduction in the demyelinated dorsal columns of
induced shape changes of astrocytes are accompanied the rat spinal cord. Journal of Neuroscience 18, 6176–6185.
by rapid depolymerization of actin. Brain Research 528, JANI, H. R. & RAISMAN, G. (2004) Ensheathing cell cul-
189–196. tures from the olfactory bulb and mucosa. Glia 47,
GOMEZ, V. M., AVERILL, S., KING, V., YANG, Q., 130–137.
P ÉREZ, E. D., CHACON, S. C., WARD, R., NIETO- KATO, T., HONMOU, O., UEDE, T., HASHI, K. &
SAMPEDRO, M., PRIESTLEY, J. & TAYLOR, J. (2003) KOCSIS, J. D. (2000) Transplantation of human olfactory
Transplantation of olfactory ensheathing cells fails to pro- ensheathing cells elicits remyelination of demyelinated
mote significant axonal regeneration from dorsal roots rat spinal cord. Glia 30, 209–218.
into the rat cervical cord. Journal of Neurocytology 32, KEY, B. & ST JOHN, J. (2002) Axon navigation in the mam-
53–70. malian primary olfactory pathway: where to next? Chem-
GONG, Q., BAILEY, M. S., PIXLEY, S. K., ENNIS, M., ical Senses 27, 245–260.
LIU, W. & SHIPLEY, M. T. (1994) Localization and reg- KUMAR, R., HAYAT, S., FELTS, P., BUNTING, S. &
ulation of low affinity nerve growth factor receptor ex- WIGLEY, C. (2005) Functional differences and interac-
pression in the rat olfactory system during development tions between phenotypic subpopulations of olfactory
and regeneration. Journal of Comparative Neurology 344, ensheathing cells in promoting CNS axonal regeneration.
336–348. Glia 50, 12–20.
GOODMAN, M. N., SILVER, J. & JACOBBERGER, J. W. LAKATOS, A., BARNETT, S. C. & FRANKLIN, R. J.
(1993) Establishment of neurite outgrowth properties of (2003) Olfactory ensheathing cells induce less host as-
neonatal and adult rat olfactory bulb glial cell lines. Brain trocyte response and chondroitin sulphate proteoglycan
Research 619, 199–213. expression than Schwann cells following transplantation
78 V I N C E N T, W E S T and C H U A H

into adult CNS white matter. Experimental Neurology 184, & AXEL, R. (1996) Visualizing an olfactory sensory map.
237–246. Cell 87, 675–686.
LAKATOS, A., FRANKLIN, R. J. & BARNETT, S. C. (2000) MORRIS, M. R., DEWITT, S., LAFFAFIAN, I. &
Olfactory ensheathing cells and Schwann cells differ in HALLETT, M. B. (2003) Phagocytosis by inflammatory
their in vitro interactions with astrocytes. Glia 32, 214–225. phagocytes: experimental strategies for stimulation and
LEE, I. H., BULTE, J. W., SCHWEINHARDT, P., quantification. Methods in Molecular Biology 225, 35–46.
DOUGLAS, T., TRIFUNOVSKI, A., HOFSTETTER, NAN, B., GETCHELL, M. L., PARTIN, J. V. &
C., OLSON, L. & SPENGER, C. (2004) In vivo mag- GETCHELL, T. V. (2001) Leukemia inhibitory factor,
netic resonance tracking of olfactory ensheathing glia interleukin-6, and their receptors are expressed tran-
grafted into the rat spinal cord. Experimental Neurology siently in the olfactory mucosa after target ablation. Jour-
187, 509–516. nal of Comparative Neurology 435, 60–77.
LI, Y., CARLSTEDT, T., BERTHOLD, C. H. & NASH, H. H., BORKE, R. C. & ANDERS, J. J. (2002)
RAISMAN, G. (2004) Interaction of transplanted Ensheathing cells and methylprednisolone promote ax-
olfactory–ensheathing cells and host astrocytic pro- onal regeneration and functional recovery in the lesioned
cesses provides a bridge for axons to regenerate across adult rat spinal cord. Journal of Neuroscience 22, 7111–
the dorsal root entry zone. Experimental Neurology 188, 7120.
300–308. NAVARRO, X., VALERO, A., GUDINO, G., FORES,
LI, Y., DECHERCHI, P. & RAISMAN, G. (2003a) Trans- J., RODRIGUEZ, F. J., VERD U , E., PASCUAL, R.,
plantation of olfactory ensheathing cells into spinal cord CUADRAS, J. & NIETO-SAMPEDRO, M. (1999) En-
lesions restores breathing and climbing. Journal of Neuro- sheathing glia transplants promote dorsal root regener-
science 23, 727–731. ation and spinal reflex restitution after multiple lumbar
LI, Y., FIELD, P. M. & RAISMAN, G. (1998) Regeneration rhizotomy. Annals of Neurology 45, 207–215.
of adult rat corticospinal axons induced by transplanted OLAND, L. A., POTT, W. M., HOWARD, C. T., INLOW,
olfactory ensheathing cells. Journal of Neuroscience 18, M. & BUCKINGHAM, J. (2003) A diffusible signal at-
10514–10524. tracts olfactory sensory axons toward their target in the
LI, Y., SAUVE, Y., LI, D., LUND, R. D. & RAISMAN, developing brain of the moth. Journal of Neurobiology 56,
G. (2003b) Transplanted olfactory ensheathing cells pro- 24–40.
mote regeneration of cut adult rat optic nerve axons. Jour- O’TOOLE, D., WEST, A. K., VICKERS, J. C. & CHUAH,
nal of Neuroscience 23, 7783–7788. M. I. (2005) Effects of olfactory ensheathing cells on as-
LIU, G. & RAO, Y. (2003) Neuronal migration from the fore- trogliosis. In: Proceedings of the Australian Neuroscience So-
brain to the olfactory bulb requires a new attractant per- ciety (edited by Paul Martin) Perth, Australia.
sistent in the olfactory bulb. Journal of Neuroscience 23, PASCUAL, J. I., GUDINO-CABRERA, G., INSAUSTI, R.
6651–6659. & NIETO-SAMPEDRO, M. (2002) Spinal implants of ol-
LIU, K. L., CHUAH, M. I. & LEE, K. K. (1995) Soluble factory ensheathing cells promote axon regeneration and
factors from the olfactory bulb attract olfactory Schwann bladder activity after bilateral lumbosacral dorsal rhizo-
cells. Journal of Neuroscience 15, 990–1000. tomy in the adult rat. Journal of Urology 167, 1522–1526.
LU, J., FERON, F., MACKAY-SIM, A. & WAITE, P. M. PASTERKAMP, R. J., DE WINTER, F., HOLTMAAT,
(2002) Olfactory ensheathing cells promote locomotor A. J. & VERHAAGEN, J. (1998) Evidence for a role
recovery after delayed transplantation into transected of the chemorepellent semaphorin III and its receptor
spinal cord. Brain 125, 14–21. neuropilin-1 in the regeneration of primary olfactory ax-
MACKAY-SIM, A. & KITTEL, P. (1991) Cell dynamics in ons. Journal of Neuroscience 18, 9962–9976.
the adult mouse olfactory epithelium: a quantitative au- P ÉREZ-BOUZA, A., WIGLEY, C. B., NACIMIENTO, W.,
toradiographic study. Journal of Neuroscience 11, 979– NOTH, J. & BROOK, G. A. (1998) Spontaneous orien-
984. tation of transplanted olfactory glia influences axonal re-
MACKAY-SIM, A. & KITTEL, P. W. (1990) On the life span generation. Neuroreport 9, 2971–2975.
of olfactory receptor neurons. European Neuroscience As- PIXLEY, S. K. (1992) The olfactory nerve contains two pop-
sociation 3, 209–215. ulations of glia, identified both in vivo and in vitro. Glia
MIRAGALL, F. & DERMIETZEL, R. (1992) Immunocyto- 5, 269–284.
chemical localization of cell adhesion molecules in the POLENTES, J., STAMEGNA, J. C., NIETO-SAMPEDRO,
developing and mature olfactory system. Microscopy Re- M. & GAUTHIER, P. (2004) Phrenic rehabilitation and
search and Technique 23, 157–172. diaphragm recovery after cervical injury and transplan-
MIRAGALL, F., KADMON, G., HUSMANN, M. & tation of olfactory ensheathing cells. Neurobiology of Dis-
SCHACHNER, M. (1988) Expression of cell adhesion ease 16, 638–653.
molecules in the olfactory system of the adult mouse: RADTKE, C., AKIYAMA, Y., BROKAW, J., LANKFORD,
presence of the embryonic form of N-CAM. Developmen- K. L., WEWETZER, K., FODOR, W. L. & KOCSIS, J.
tal Biology 129, 516–531. D. (2004) Remyelination of the nonhuman primate spinal
MIRAGALL, F., KADMON, G. & SCHACHNER, M. (1989) cord by transplantation of H-transferase transgenic adult
Expression of L1 and N-CAM cell adhesion molecules pig olfactory ensheathing cells. The FASEB Journal 18,
during development of the mouse olfactory system. De- 335–337.
velopmental Biology 135, 272–286. RAISMAN, G. (2001) Olfactory ensheathing cells - another
MOMBAERTS, P., WANG, F., DULAC, C., CHAO, S. K., miracle cure for spinal cord injury? Nature Reviews Neu-
NEMES, A., MENDELSOHN, M., EDMONDSON, J. roscience 2, 369–375.
Morphological and functional plasticity of olfactory ensheathing cells 79

RAMAKERS, G. J. & MOOLENAAR, W. H. (1998) Regula- tral nervous system injury in rats and humans. Journal of
tion of astrocyte morphology by RhoA and lysophos- Neurotrauma 18, 377–388.
phatidic acid. Experimental Cell Research 245, 252– SCHWARTING, G. A., KOSTEK, C., AHMAD, N.,
262. DIBBLE, C., PAYS, L. & PUSCHEL, A. W. (2000)
RAMER, L. M., RICHTER, M. W., ROSKAMS, A. Semaphorin 3A is required for guidance of olfactory ax-
J., TETZLAFF, W. & RAMER, M. S. (2004a) ons in mice. Journal of Neuroscience 20, 7691–7697.
Peripherally–derived olfactory ensheathing cells do not SCOTTI, A. L., HOFFMANN, M. C. & NITSCH, C. (1994)
promote primary afferent regeneration following dorsal The neurite growth promoting protease nexin 1 in glial
root injury. Glia 47, 189–206. cells of the olfactory bulb of the gerbil: an ultrastructural
RAMER, L. M., AU, E., RICHTER, M. W., LIU, J., study. Cell and Tissue Research 278, 409–413.
TETZLAFF, W. & ROSKAMS, A. J. (2004b) Peripheral SMALE, K. A., DOUCETTE, R. & KAWAJA, M. D. (1996)
olfactory ensheathing cells reduce scar and cavity forma- Implantation of olfactory ensheathing cells in the adult
tion and promote regeneration after spinal cord injury. rat brain following fimbria-fornix transection. Experimen-
Journal of Comparative Neurology 473, 1–15. tal Neurology 137, 225–233.
RAM O N-CUETO, A., CORDERO, M. I., SANTOS- SMITH, P. M., LAKATOS, A., BARNETT, S. C., JEFFERY,
BENITO, F. F. & AVILA, J. (2000) Functional recovery N. D. & FRANKLIN, R. J. (2002) Cryopreserved cells
of paraplegic rats and motor axon regeneration in their isolated from the adult canine olfactory bulb are ca-
spinal cords by olfactory ensheathing glia. Neuron 25, pable of extensive remyelination following transplanta-
425–435. tion into the adult rat CNS. Experimental Neurology 176,
RAM O N-CUETO, A. & NIETO-SAMPEDRO, M. (1994) 402–406.
Regeneration into the spinal cord of transected dorsal SMITH, P. M., SIM, F. J., BARNETT, S. C. & FRANKLIN,
root axons is promoted by ensheathing glia transplants. R. J. (2001) SCIP/Oct-6, Krox-20, and desert hedgehog
Experimental Neurology 127, 232–244. mRNA expression during CNS remyelination by trans-
RAM ÓN-CUETO, A. , P ÉREZ, J. & NIETO-SAMPEDRO, planted olfactory ensheathing cells. Glia 36, 342–353.
M. (1993) In vitro enfolding of olfactory neurites by SONIGRA, R. J., BRIGHTON, P. C., JACOBY, J., HALL,
p75 NG F receptor positive ensheathing cells from adult S. & WIGLEY, C. B. (1999) Adult rat olfactory nerve
rat olfactory bulb. European Journal of Neuroscience 5, ensheathing cells are effective promoters of adult central
1172–1180. nervous system neurite outgrowth in coculture. Glia 25,
RAM ÓN-CUETO, A., PLANT, G. W., AVILA, J. & 256–269.
BUNGE, M. B. (1998) Long-distance axonal regenera- ST JOHN, J. A., TISAY, K. T., CARAS, I. W. & KEY, B.
tion in the transected adult rat spinal cord is promoted (2000) Expression of EphA5 during development of the
by olfactory ensheathing glia transplants. Journal of Neu- olfactory nerve pathway in rat. Journal of Comparative
roscience 18, 3803–3815. Neurology 416, 540–550.
RAM ÓN-CUETO, A. & VALVERDE, F. (1995) Olfactory STORAN, M. J., MAGNALDO, T., BIOL-N’GARAGBA,
bulb ensheathing glia: a unique cell type with axonal M. C., ZICK, Y. & KEY, B. (2004) Expression and puta-
growth- promoting properties. Glia 14, 163–173. tive role of lactoseries carbohydrates present on NCAM
REINHARD, E., MEIER, R., HALFTER, W., ROVELLI, G. in the rat primary olfactory pathway. Journal of Compara-
& MONARD, D. (1988) Detection of glia-derived nexin tive Neurology 475, 289–302.
in the olfactory system of the rat. Neuron 1, 387–394. SUIDAN, H. S., NOBES, C. D., HALL, A. & MONARD, D.
RIDDELL, J. S., ENRIQUEZ-DENTON, M., TOFT, A., (1997) Astrocyte spreading in response to thrombin and
FAIRLESS, R. & BARNETT, S. C. (2004) Olfactory en- lysophosphatidic acid is dependent on the Rho GTPase.
sheathing cell grafts have minimal influence on regener- Glia 21, 244–252.
ation at the dorsal root entry zone following rhizotomy. SURVEYOR, G. A. & BRIGSTOCK, D. R. (1999) Immuno-
Glia 47, 150–167. histochemical localization of connective tissue growth
RIDLEY, A. J. & HALL, A. (1992) The small GTP-binding factor (CTGF) in the mouse embryo between days 7.5
protein rho regulates the assembly of focal adhesions and and 14.5 of gestation. Growth Factors 17, 115–124.
actin stress fibers in response to growth factors. Cell 70, SUZUKI, Y., SCHAFER, J. & FARBMAN, A. I. (1995)
389–399. Phagocytic cells in the rat olfactory epithelium after bul-
ROSSLER, W., OLAND, L. A., HIGGINS, M. R., bectomy. Experimental Neurology 136, 225–233.
HILDEBRAND, J. G. & TOLBERT, L. P. (1999) Devel- SUZUKI, Y., TAKEDA, M. & FARBMAN, A. I. (1996) Sup-
opment of a glia-rich axon-sorting zone in the olfactory porting cells as phagocytes in the olfactory epithelium
pathway of the moth Manduca sexta. Journal of Neuro- after bulbectomy. Journal of Comparative Neurology 376,
science 19, 9865–9877. 509–517.
SAFAVI-ABBASI, S., WOLFF, J. R. & MISSLER, M. (2001) TAKAMI, T., OUDEGA, M., BATES, M. L., WOOD, P. M.,
Rapid morphological changes in astrocytes are accompa- KLEITMAN, N. & BUNGE, M. B. (2002) Schwann cell
nied by redistribution but not by quantitative changes of but not olfactory ensheathing glia transplants improve
cytoskeletal proteins. Glia 36, 102–115. hindlimb locomotor performance in the moderately con-
SCHWAB, J. M., BESCHORNER, R., NGUYEN, T. D., tused adult rat thoracic spinal cord. Journal of Neuroscience
MEYERMANN, R. & SCHLUESENER, H. J. (2001) 22, 6670–6681.
Differential cellular accumulation of connective tissue TAYLOR, J. S., MUNETON-GOMEZ, V. C., EGUIA-
growth factor defines a subset of reactive astrocytes, in- RECUERO, R. & NIETO-SAMPEDRO, M. (2001)
vading fibroblasts, and endothelial cells following cen- Transplants of olfactory bulb ensheathing cells promote
80 V I N C E N T, W E S T and C H U A H

functional repair of multiple dorsal rhizotomy. Progress E. (2003) Pneumococcal carriage results in ganglioside-
in Brain Research 132, 641–654. mediated olfactory tissue infection. Proceedings of the Na-
THORNE, R. G., PRONK, G. J., PADMANABHAN, V. tional Academy of Science 100, 14363–14367.
& FREY, W. H., 2ND (2004) Delivery of insulin-like VERD U , E., NAVARRO, X., GUDINO-CABRERA, G.,
growth factor-I to the rat brain and spinal cord along RODRIGUEZ, F. J., CEBALLOS, D., VALERO, A.
olfactory and trigeminal pathways following intranasal & NIETO-SAMPEDRO, M. (1999) Olfactory bulb en-
administration. Neuroscience 127, 481–496. sheathing cells enhance peripheral nerve regeneration.
TOEWS, A. D., BARRETT, C. & MORELL, P. (1998) Neuroreport 10, 1097–1101.
Monocyte chemoattractant protein 1 is responsible for VICKLAND, H., WESTRUM, L. E., KOTT, J. N.,
macrophage recruitment following injury to sciatic PATTERSON, S. L. & BOTHWELL, M. A. (1991) Nerve
nerve. Journal of Neuroscience Research 53, 260–267. growth factor receptor expression in the young and adult
TRAN, P. B. & MILLER, R. J. (2003) Chemokine receptors: rat olfactory system. Brain Research 565, 269–279.
signposts to brain development and disease. Nature Re- VINCENT, A. J., TAYLOR, J. M., CHOI-LUNDBERG, D.
views Neuroscience 4, 444–455. L., WEST, A. K. & CHUAH, M. I. (2005) Genetic expres-
TRELOAR, H. B., PURCELL, A. L. & GREER, C. A. (1999) sion profile of olfactory ensheathing cells is distinct from
Glomerular formation in the developing rat olfactory that of Schwann cells and astrocytes. Glia 51, 132–147.
bulb. Journal of Comparative Neurology 413, 289–304. VINCENT, A. J., WEST, A. K., CHOI-LUNDBERG, D. L.
TURNER, C. P. & PEREZ-POLO, J. R. (1992) Regulation & CHUAH, M. I. (2005) Olfactory ensheathing cells dis-
of the low affinity receptor for nerve growth factor, play antimicrobial activity in culture and may contribute
p75NGFR, in the olfactory system of neonatal and adult to the innate immune defense of the olfactory system. In:
rat. International Journal of Developmental Neuroscience 10, Proceedings of the Australian Neuroscience Society (edited
343–359. by Paul Martin) Perth, Australia.
UBINK, R., HALASZ, N., ZHANG, X., DAGERLIND, A. VINCENT, A. J., WEST, A. K. & CHUAH, M. I. (2003)
& H ÖKFELT, T. (1994) Neuropeptide tyrosine is ex- Morphological plasticity of olfactory ensheathing cells is
pressed in ensheathing cells around the olfactory nerves regulated by cAMP and endothelin-1. Glia 41, 393–403.
in the rat olfactory bulb. Neuroscience 60, 709–726. WEWETZER, K., KERN, N., EBEL, C., RADTKE, C. &
UBINK, R. & H ÖKFELT, T. (2000) Expression of neuropep- BRANDES, G. (2005) Phagocytosis of O4+ axonal frag-
tide Y in olfactory ensheathing cells during prenatal de- ments in vitro by p75- neonatal rat olfactory ensheathing
velopment. Journal of Comparative Neurology 423, 13–25. cells. Glia 49, 577–587.
VALVERDE, F., SANTACANA, M. & HEREDIA, M. WEWETZER, K., VERD U , E., ANGELOV, D. N. &
(1992) Formation of an olfactory glomerulus: morpho- NAVARRO, X. (2002) Olfactory ensheathing glia and
logical aspects of development and organization. Neuro- Schwann cells: two of a kind? Cell and Tissue Research
science 49, 255–275. 309, 337–345.
VAN DEN POL, A. N. & SANTARELLI, J. G. (2003) Ol- WHITESIDES, J. G., 3RD & LAMANTIA, A. S. (1996) Dif-
factory ensheathing cells: time lapse imaging of cellular ferential adhesion and the initial assembly of the mam-
interactions, axonal support, rapid morphological shifts, malian olfactory nerve. Journal of Comparative Neurology
and mitosis. Journal of Comparative Neurology 458, 175– 373, 240–254.
194. YAN, H., LU, D. & RIVKEES, S. A. (2003) Lysophospha-
VAN GINKEL, F. W., MCGHEE, J. R., WATT, J. M., tidic acid regulates the proliferation and migration of ol-
CAMPOS-TORRES, A., PARISH, L. A. & BRILES, D. factory ensheathing cells in vitro. Glia 44, 26–36.

View publication stats

You might also like