Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Mycobacterium leprae-induced demyelination: a model for

early nerve degeneration


Anura Rambukkana

The molecular events that occur at the early phase of many specialized organelle, which forms a multi-lamellar struc-
demyelinating neurodegenerative diseases are unknown. A recent ture around axons [1]. Myelin is produced by oligoden-
demonstration of rapid demyelination and axonal injury induced drocytes and Schwann cells, supporting glial cells of the
by Mycobacterium leprae provides a model for elucidating the central and peripheral nervous systems (CNS and PNS),
molecular events of early nerve degeneration which might be respectively [1–3]. In myelinated nerve fibers, myelin is
common to neurodegenerative diseases of both infectious origin arranged in segments separated by the nodes of Ranvier,
and unknown etiology. The identification of the M. leprae-targeted where voltage-gated sodium channels are clustered in
Schwann cell receptor, dystroglycan, and its associated high density in the axon membrane so that they can
molecules in myelination, demyelination and axonal functions produce action potentials via saltatory conduction (the
suggests a role for these molecules in early nerve degeneration. action potentials jump from node to node; [4–6]). The
bulk of the myelin sheath is formed of compact myelin,
Addresses which covers and masks the internodal areas of the axon
The Rockefeller University, Bronk building, Room 501, comprised of fewer sodium channels and a higher density
1230 York Avenue, New York, New York 10021, USA of potassium channels, which tends to oppose the gen-
e-mail: rambuka@imap.rockefeller.edu
eration of action potentials [4–6]. The crucial role of
myelination of axons in nervous system functions is
Current Opinion in Immunology 2004, 16:511–518 evident from the large number of neurological diseases
that occur as a result of demyelination.
This review comes from a themed issue on
Host–pathogen interactions
Edited by Tom Ottenhoff and Michael Bevan Demyelination is a common pathological feature of
neurodegenerative diseases of both infectious origin, such
Available online 15th June 2004 as leprosy, and unknown etiology, such as multiple sclero-
0952-7915/$ – see front matter sis (MS) and Guillain-Barre syndrome (GBS) in the CNS
ß 2004 Elsevier Ltd. All rights reserved. and PNS, respectively [7–11]. One of the classical exam-
ples of infectious neurodegenerative diseases with per-
DOI 10.1016/j.coi.2004.05.021 ipheral nerve degeneration/neuropathies is leprosy,
which is caused by the non-toxic/non-cytolytic bacterium
Abbreviations Mycobacterium leprae [7]. Nerve damage caused by leprosy
CNS central nervous system comprises both demyelinating neuropathies and axonal
DRP2 dystroglycan-related protein 2 neuropathies [7,8]. Whereas the former causes deleter-
G globular
GBS Guillain-Barre syndrome
ious effects that originate primarily in myelinating
LCMV lymphocytic choriomeningitis virus Schwann cells, in the latter, neurons (axons), particularly
LL lepromatous leprosy sensory neurons, are initially affected. Damage to the
MS multiple sclerosis myelin sheath is accompanied by a decrease in action-
PGL-1 phenolic glycolipid-1
potential conduction velocity, a physiological hallmark of
PNS peripheral nervous system
demyelination [2,9]. In the case of MS, the diagnosis rests
in part on the demonstration of such conduction velocity
Introduction [2,9]. Although demyelinating diseases are generally
An important challenge in therapeutic interventions for thought of as those in which myelin is primarily affected,
demyelinating neurodegenerative diseases is to under- subtle changes in the axon may also have profound effects
stand the cellular and molecular events that underlie the on glial cell function, myelin maintenance and subse-
different phases of the disease. However, the molecular quent remyelination [9,10]. Therefore, similar to the
events that occur during the early phase of neurodegen- impairment seen in MS sufferers, neurological impair-
erative diseases are unknown, mainly because the etiol- ment in many other demyelinating diseases may also be
ogy or the trigger that initiates the disease processes have associated with axonal degeneration [10]. Interestingly,
not been identified and, therefore, it is almost impossible somewhat similar pathological features have also been
to dissect the underlying mechanisms of the early phase observed in leprosy patients with peripheral nerve
of these diseases. damage [7,8]. Disabilities as a result of nerve damage
may be partly due to such M. leprae-induced demyelina-
In the mammalian nervous system, the rapid conduction tion and axonal injuries. A correlation between inflam-
of action potentials depends on the myelin sheath, a matory reactions and demyelination/axonal damage has

www.sciencedirect.com Current Opinion in Immunology 2004, 16:511–518


512 Host–pathogen interactions

been observed in MS, GBS and leprosy [8,10,11]. molecules, phenolic glycolipid-1 (PGL-1; [13]) and ML-
However, it is largely unknown whether other non- LBP21 (for M. leprae laminin-binding 21 kDa protein;
immune-mediated mechanisms are involved in myelin [14]) have been identified as key bacterial molecules.
damage and axonal degeneration in these diseases. The corresponding key host molecules are the Schwann
cell basal laminae component laminin-2 [15] and its
This review analyzes the potential mechanisms involved receptors dystroglycan [16] and b4 integrin [15]. Consis-
in M. leprae-induced demyelination as a model represent- tent with the capacity of the M. leprae cell-wall fraction to
ing the early phase of nerve degeneration, and how induce demyelination, it was shown that PGL-1 in pur-
M. leprae may use such an early nerve injury response ified form could also induce demyelination as effectively
as a strategy for intracellular survival. as the whole cell wall fraction [12]. Because PGL-1
interacts with the globular (G) domain of laminin-2,
Common themes at the onset of particularly the G4-5 region, which is known to bind to
demyelination: M. leprae as a model a-dystroglycan [17], it is possible that activation of signal-
As demyelination is a common neuropathological feature ing in myelinated Schwann cells by M. leprae or PGL-1 via
of leprosy, MS and GBS, and there are certain structural the laminin-2-dystroglycan pathway may contribute to
and functional similarities between the organization of the breakdown of the myelin sheath as a result of dereg-
the myelinated axons of the CNS and PNS [4,5], it is ulation of the signaling network that maintains the
reasonable to believe that there may be a common theme mature myelin sheaths.
at the onset of demyelination. Neurotrophic pathogens,
particularly those that do not cause direct cell lysis or Role of laminin-2 and its receptors in
cytopathic effect, can be used as models in dissecting myelination and demyelination
early events of demyelination. In myelinated nerve fibers, laminin-2 is the major com-
ponent of the basal lamina/extracellular matrix, which
Recent studies have shown that both demyelination and deposits just outside Schwann cell membrane (see
axonal injury can occur at the onset of M. leprae infection Figure 1). During development, Schwann cells myelinate
in the absence of immune cells [12]. Although immune axons in the continuous presence of basal lamina [3,18].
responses play a critical role in the clinical manifestation In order for the Schwann cell to respond to axonal signals
of the disease, the identification of non-immune- to initiate myelination, the Schwann cell must synthesize
mediated demyelination induced by M. leprae proposes extracellular matrix molecules (mainly the laminin-2 iso-
the existence of alternative mechanism(s) for demyelina- form and type IV collagen) and assemble them into the
tion. These studies have shown that the binding of basal lamina [18–20]. Under conditions in which the basal
M. leprae to myelinated Schwann cell axon units is suffi- lamina fails to assemble, Schwann cells are unable to
cient to induce demyelination without causing apoptosis myelinate normally [18]. The genetic evidence for the
or cell death of Schwann cells or neurons [12]. Thus, it is role of laminin-2 in myelination is based on the studies of
likely that perturbation of homeostasis of the neural dystrophic mice and humans with congenital muscular
microenvironment, particularly the deregulation of exist- dystrophy (CMD); both have mutations in the laminin-a2
ing signaling networks that maintain the integrity of gene and are characterized by defects in Schwann-cell
myelin sheaths in mature Schwann cells, may be respon- myelination [21].
sible for rapid demyelination.
Laminin-2 in the basal lamina anchors to Schwann cells
Although such early demyelination may not be sufficient via laminin-2 receptors, such as a6b1 integrin, a6b4
to exert a clinical manifestation of the disease, the under- integrin and dystroglycan [22,23]. Despite the known
lying molecular basis may provide information for devel- role of laminin-2 in myelination, the mechanism of
oping early diagnostic and therapeutic intervention action of laminin-2 and the role of laminin-2 receptors
before the disease is aggravated by immune attack. in myelination are largely unknown. Findings by Feltri
et al. [24] provide important insights into the role of
Role of M. leprae cell wall components and laminin-2 receptor b1 integrin in the differentiation of
Schwann cell receptors myelinating Schwann cells. If the b1 integrin gene is
As the binding of M. leprae and M. leprae cell wall conditionally deleted in immature Schwann cells, the
components to myelinated Schwann cell axon units is myelination process is markedly delayed and even-
sufficient to induce demyelination, the possibility exists tually leads to the development of progressive neuro-
that receptors in myelinated Schwann cells that are asso- pathy [24]. Although there is no evidence for the
ciated with M. leprae interactions may play a role in interaction of M. leprae with b1 integrin, M. leprae
perturbation of the homeostasis of the neural microenvir- may interfere with existing crosstalk between the lami-
onment. Recent studies have shown a mechanism by nin receptors (or non-laminin receptors) responsible for
which M. leprae interacts with myelinated Schwann cells; maintaining the mature myelin sheaths and mediating
two of the most important M. leprae cell-wall-associated cell signaling.

Current Opinion in Immunology 2004, 16:511–518 www.sciencedirect.com


M. leprae-induced demyelination Rambukkana 513

Figure 1

M. leprae M. leprae
(a) (b)
Nucleus

Receptors
Axons Myelin
Axon sheath

Laminin-2
Basal lamina

Non-myelinated Schwann cell Myelinated Schwann cell

M. leprae
M. leprae Resist
invasion M. leprae
invasion

(c) Multiplication (d)


and release -Demyelination
-Axonal damage
(Nerve injury responses)
Proliferation
Re-infection

Proliferation
Innate and adaptive
Generate more immune responses
Schwann cells

Nerve regeneration

Bacterial survival

Nerve degeneration

Current Opinion in Immunology

Mycobacterium leprae-induced early demyelination and nerve injury response as both a bacterial strategy for intracellular survival and a
trigger for initiating immune-mediated response and injury. Upon PNS infection, M. leprae attaches to both (a) non-myelinated and (b)
myelinated Schwann cells, but preferentially invades the non-myelinated phenotype, where it multiplies, releases and re-infects more
non-myelinated Schwann cells. M. leprae attachment to myelinated Schwann cells is sufficient to induce demyelination and axonal damage.
To continue the intracellular life cycle within its preferred niche and to establish persistent infection, M. leprae generates increasing numbers
of non-myelinated Schwann cells both (c) directly by inducing proliferation of non-myelinated Schwann cells, and (d) indirectly by causing nerve
injury, demyelination and axonal damage, which normally induces Schwann cell proliferation to repair the damaged nerve (nerve regeneration).
Early nerve injury might also cause the destabilization of the neural microenvironment, leading to a cascade of other cellular responses that
eventually recruit the immune cells of the innate and adaptive immune systems, causing lasting damage to peripheral nerves.

Role of dystroglycan in myelination and is a Schwann cell receptor for non-lytic neurotrophic
demyelination pathogens, such as M. leprae and lymphocytic choriome-
An important non-integrin laminin-2 receptor in Schwann ningitis virus (LCMV), and Lassa fever virus of the
cells is dystroglycan, which is encoded by a single gene arenavirus family [16,26,27]. Dystroglycan thought to
and cleaved into two proteins, a- and b-dystroglycan be involved in the initiation of myelination, as dystrogly-
[23,25]. Recent studies have shown that a-dystroglycan can is overexpressed with laminin-2 during peripheral

www.sciencedirect.com Current Opinion in Immunology 2004, 16:511–518


514 Host–pathogen interactions

nerve myelination in postnatal rats [28], and its expression contains two PDZ domains and is implicated in the
is regulated in parallel with laminin-2 throughout the assembly of macromolecular signaling complexes, how-
degeneration process after nerve crush injury [29]. ever, is exquisitely Schwann-cell specific, and is concen-
Furthermore, recent studies have shown that leprosy trated to the abaxonal plasma membrane in the mature
bacillus and its component PGL-1 [30], which bind to myelin sheath [33]. Periaxin is essential for the stability of
dystroglycan on Schwann cells through the G domain of myelin sheaths, as mice and humans lacking periaxin
laminin-2 [13], cause demyelination [12]. develop a demyelinating peripheral neuropathy, which
manifests as a mixture of hypermyelination and demye-
Direct genetic evidence for the involvement of dystro- lination [33]. The late onset of demyelination in periaxin-
glycan in peripheral nerve function has recently been null mice suggests that the dystroglycan–DRP2–periaxin
demonstrated by Saito et al. [31] who showed that complex has a role in maintaining the myelin sheath,
selective deletion of Schwann cell dystroglycan (null possibly via signal transduction [33,34].
P0 mice lacking dystroglycan) results in severe neurolog-
ical dysfunction and a broad spectrum of morphological Because contact with the basal lamina modulates the
abnormalities in myelination and nodal architecture, lead- expression of genes required for myelination, laminin-2
ing to defects in nerve conduction. Importantly, these receptors and associated molecules have been implicated
mice have dispersed distribution of sodium ion channels in mediating the responsiveness of Schwann cells to
on the node of Ranvier, suggesting that disruption of the extracellular signals from laminin-2 [35,36]. Such signals
laminin–dystroglycan interaction is crucial not only for could be induced by M. leprae upon attachment to lami-
peripheral nerve myelination but also for the functions of nin-2 in the basal lamina. Periaxin binds DRP2 and
ion channels at the nodes of myelinated axons [31]. dimerizes at its PDZ domain, which probably contributes
However, mechanisms by which dystroglycan regulates to the distinctive clustering of the complex in the myelin-
the myelination process and ion-channel expression is forming Schwann-cell membrane [33]. Therefore, signal-
unknown. As M. leprae interacts with Schwann cells via ing through the dystroglycan–DRP2–periaxin complex
dystroglycan and laminin-2, and dystroglycan is involved may regulate the maintenance of the myelin sheath
in signal transduction and myelination [25], signaling during myelination, and M. leprae binding to laminin-2
through dystroglycan is likely to play a key role in may profoundly affect signaling mediated through the
M. leprae-induced demyelination and axonal degeneration dystroglycan–DRP2–periaxin complex, thereby promot-
during the early phase of infection. ing breakdown of the myelin sheaths. As M. leprae appears
to use multiple strategies to subvert Schwann cell func-
Similar to M. leprae, LCMV uses dystroglycan as a recep- tions [37], one cannot exclude the possibility of involve-
tor for viral entry [27], but does not interact with laminin- ment of other non-laminin receptors in M. leprae
2 for Schwann-cell interactions [26]. Unlike M. leprae, interactions with myelinated Schwann cells, and signaling
LCMV infection does not cause any apparent effect on mediated through such receptors could also be respon-
mature myelinating Schwann cells [12,26]. However, sible in mediating demyelination.
persistent infection of immature Schwann cells rendered
these cells defective or incapable of forming a myelin Early demyelination/nerve injury response as
sheath when they differentiate into a myelinating phe- a bacterial survival strategy
notype in an in vitro differentiation model of Schwann Non-myelinating Schwann cells as preferential targets
cells [26]. Defects in myelination coincide with the for M. leprae invasion
downregulation of dystroglycan protein expression and In the PNS, Schwann cells exist in two phenotypes,
disruption of the assembly of the laminin-2 network and myelinating Schwann cells that form one-to-one relation-
basal lamina on Schwann cell axon units. In addition, ships with large caliber axons and non-myelinating
infected Schwann cells that form mature myelin sheaths Schwann cells that ensheath multiple small axons
undergo demyelination. These results further underscore (Figure 1; [3]). The former propagates rapid nerve con-
the role of the laminin–dystroglcan linkage in myelina- duction due to the larger caliber axon and the myelin
tion and demyelination processes, and show the potential sheath, whereas the latter is mostly comprises smaller
use of LCMV as a model to dissect the molecular basis of sensory axons and provides slow nerve conduction.
myelination. Recent studies have shown that myelinated and non-
myelinated Schwann cells show distinct functional
Role of the dystroglycan–DRP2–periaxin responses to M. leprae infection. Although M. leprae binds
complex in myelination and demyelination to both myelinated and non-myelinated Schwann cell-
In myelinating Schwann cells, dystroglycan forms a com- axon units (Figure 1; [15]), it is the non-myelinated
plex with dystroglycan-related protein 2 (DRP2) and Schwann cells that are susceptible to M. leprae invasion
periaxin [32,33]. DRP2 is highly enriched in Schwann and preferentially harbor M. leprae in the PNS [12].
cells and is specifically localized to the cytoplasm that Therefore, non-myelinating Schwann cells are not only
directly opposes the myelin sheath [33]. Periaxin, which the natural host for the multiplication of M. leprae but also

Current Opinion in Immunology 2004, 16:511–518 www.sciencedirect.com


M. leprae-induced demyelination Rambukkana 515

protect the organism from host immune responses Figure 2


[7,8,12,37,38]. For this reason, non-myelinating Schwann
cell resident M. leprae is a primary source of infection that (a)
not only causes nerve injury but is also responsible for the
continuous leakage of M. leprae and its antigens into the M
neural tissue milieu and circulation, and thus may lead to
a state of persistent infection or relapse (Figure 1; [8,38]). Ax
Myelinated Schwann cells, however, are extremely resis-
tant to M. leprae invasion but undergo demyelination and
axonal damage upon attachment to Schwann cell axon
units. In vivo induction of demyelination in lymphocyte-
deficient Rag-1/ mice [12] is also caused by similar
mechanism, as M. leprae fails to invade Schwann cells in
mice in general, but instead induces immune-mediated
demyelination in wild-type mice as a late event [39]. The
resistance of myelinated Schwann cells to M. leprae inva-
sion in in vitro cultures may be due to the compact myelin
sheath that physically occupies almost the entire cyto- (b)
plasm of this phenotype (Figure 2a). Interestingly, early
histopathological studies of nerve biopsies from leproma- M
tous leprosy (LL) patients have also shown that intracel- Ax
lular M. leprae is frequently seen in non-myelinated
Schwann cells in the early stage, but bacilli are rarely
seen in myelinating Schwann cells even in advanced LL
patients with high bacterial load [40]. These data rein-
force recent in vitro finding that demyelination during
early infection, before the immune response comes into
play, may be caused solely by the interaction of M. leprae
or its products with myelinating Schwann cells.

Nerve injury response as means of propagating


Schwann cells/intracellular niche
The cellular preference of the non-myelinating pheno- Current Opinion in Immunology
type may provide M. leprae with a clear advantage for its
intracellular survival within the PNS. Because M. leprae Early in vivo demyelination induced by Mycobacterium leprae cell
induces both demyelination and axonal damage in vitro, wall in the absence of an adaptive immune response. The electron
micrographs are representative of sciatic nerves from Rag-1/ mice,
this condition is somewhat similar to peripheral nerve
which lack T and B cells, 72 hours after intraneural administration of
injury in vivo, where Schwann cells rapidly proliferate and (a) phosphate-buffered saline and (b) M. leprae cell wall.
enclose the injured axons, an important event for the Demyelinated and myelinated Schwann-cell axon units are shown by
promotion of nerve regeneration [41]. Strikingly, the arrows and arrowheads, respectively. Ax, axon; M, myelin sheath.
Schwann cells proliferate significantly in M. leprae-treated
nerve tissue cultures with substantial demyelination and
axonal damage [12]. Such a neural microenvironment generate more Schwann cells to secure it’s intracellular
created by early M. leprae infection may contribute to niche. Recent studies provide evidence that M. leprae
Schwann cell proliferation. Therefore, it is likely that, generate increasing number of Schwann cells for long-
once invaded, M. leprae multiply as long as non-myelinat- term intracellular survival by inducing nerve injury in
ing Schwann cells can tolerate the bacterial load (due to myelinated Schwann cells so that newly formed Schwann
the lack of anti-microbicidal machinery in Schwann cells) cells (which usually lack myelin sheath) can be further
and then release and invade more non-myelinating infected by released bacteria (Figure 1). It appears that
Schwann cells or bind myelinated Schwann cells that sub- M. leprae takes full advantage of the property of peripheral
sequently induce nerve degeneration (Figure 1). Be- nerve injury response to propagate the bacterial reservoir.
cause M. leprae is an obligate intracellular bacterium — Interestingly, more recent data show that the M. leprae
it must invade its preferred non-myelinating phenotype that reside inside Schwann cells can also induce cell
for survival. However, as the infection progresses and proliferation (N Tapinos and A Rambukkana, unpub-
bacteria undergo unrestrained multiplication, the avail- lished). This provides an alternative strategy for pro-
ability of non-myelinating Schwann cells becomes a limit- pagating the intracellular niche for bacterial survival
ing factor. To avoid such a situation, leprosy bacilli must (Figure 1). By inducing demyelination and axonal

www.sciencedirect.com Current Opinion in Immunology 2004, 16:511–518


516 Host–pathogen interactions

damage in myelinating Schwann cells from the outside, of neurological injury and initiation of the disease process,
and by directing the proliferation of non-myelinating it is the immune-mediated nerve damage that eventually
Schwann cells from inside the cells, M. leprae seems to manifests clinically [8]. One of the earliest clinical symp-
have evolved to propagate its preferred niche for long- toms of leprosy is the sensory loss, which is thought to be
term intracellular survival. caused by immune-mediated responses against M. leprae
antigens released from infected Schwann cells. Sensory
Is early demyelination a trigger for axons are mostly enclosed by non-myelinating Schwann
immune-mediated responses? cells, in which M. leprae preferentially reside for long
Although nerve degeneration in the early phase of periods of time. When M. leprae and M. leprae antigens are
M. leprae infection both in vitro and in vivo does not released from such non-myelinating Schwann cells, both
involve immune cells, such nerve injury is likely to cause non-myelinating and myelinating Schwann cells are
the destabilization of the neural microenvironment. This subjected to immune attack by infiltrating macrophages,
might subsequently lead to a cascade of other cellular T cells and the cytokines released from these inflamma-
responses that eventually recruit immune cells, particu- tory cells [7,8]. The outcome of such inflammatory
larly the cells of the innate immune system. Evidence responses is the destruction of both Schwann cell phe-
from studies with Rag-1/ knockout mice, which lack notypes (comprising functional peripheral nerves) and
mature B and T cells, and are thus unable to mount subsequent sensorimotor loss. Studies with cultured
adaptive immune response, showed the induction of human Schwann cells have shown this possibility, as
significant demyelination in sciatic nerves three days after Schwann cells process and present M. leprae, as well as
intraneural administration of M. leprae and its cell-wall recombinant proteins and peptides to MHC-class-II-
fraction (Figure 2; [12]). Although such early demyelina- restricted CD4þ T cells, and are efficiently killed by
tion in Rag-1/ mice is mediated in the absence of B and these activated T cells [46]. These findings provide
T cells, more recent studies provide evidence that the experimental evidence for a potential immune-mediated
components of the innate immune response come into mechanism of nerve damage in leprosy.
play during the late events of the infectious process
(N Tapinos and A Rambukkana, unpublished). Conclusions
Nerve damage, the hallmark of M. leprae infection,
One of the families of pattern-recognition receptors of the appears to occur during all stages of infection, although
innate immune system is the Toll-like receptor (TLR) early neurological abnormalities are not clinically detect-
family, which have been implicated in the initiation of able. Recent data suggest that early demyelination pro-
CD4þ T-cell responses [42]. Recent studies have identi- vides M. leprae with a survival advantage and possibly
fied the expression of TLRs in human leprosy skin aids the progression of infection, as demyelination and
lesions, and it has been suggested that regulated expres- subsequent axonal degeneration induce Schwann cell
sion and activation of TLRs at the site of disease con- proliferation, which, in turn, creates abundant non-
tribute to host defense against M. leprae [43]. More myelinating-type Schwann cells for M. leprae invasion,
recently, Oliveira et al. [44] have shown the expression bacterial replication and subsequent disease progression.
of TLR2 receptors on human Schwann cells. As TLRs are Understanding the different phases of M. leprae-induced
known to activate adaptive immune responses [42], it is neurodegeneration, in which non-immune, innate
possible that TLRs contribute to the development of immune and adaptive immune responses play key roles,
initial neurological injury in vivo following non-immune- should provide the rational for developing therapeutic
mediated demyelination and axonal damage. interventions not only for leprosy but also for demyeli-
nating neurodegenerative diseases in general. Therefore,
When cells of the innate immune response are at the site future studies should be directed towards delineating the
of lesion, they recognize and respond to M. leprae, and cellular and molecular basis of the different phases of
then activate adaptive immune responses by releasing demyelination and axonal damage during M. leprae infec-
inflammatory mediators that mobilize neutrophils and tion. Importantly, elucidating the molecular basis of
macrophages. As neutrophils, macrophages and pro- strategies that M. leprae utilize to survive and establish
inflammatory cytokines have been implicated in demye- a productive infection within the PNS will provide new
linating diseases [45], it is likely that the sequential insights into the basic biology of glial cells.
recruitment and propagation of immune cell populations,
preceded by non-immune-mediated demyelination and
axonal damage at the site of infection, could eventually Acknowledgements
I thank all of our collaborators and the members of my laboratory for
cause further aggravation of neurological injury during the their contribution to the original published work, from which this review
course of an M. leprae infection. was made possible. The studies cited from the author’s laboratory were
supported by World Health Organization and RO1 grants AI45816 and
NS45187 from the National Institute of Allergy and Infectious Diseases
It should be emphasized that, although non-immune- (NIAID) and The National Institute of Neurological Disorders and
mediated mechanisms might play a role in the early phase Stroke (NINDS).

Current Opinion in Immunology 2004, 16:511–518 www.sciencedirect.com


M. leprae-induced demyelination Rambukkana 517

References and recommended reading 21. Matsumura K, Yamada H, Saito F, Sunada Y, Shimizu T: Peripheral
nerve involvement in merosin-deficient congenital muscular
Papers of particular interest, published within the annual period of dystrophy and dy mouse. Neuromuscul Disord 1997, 7:7-12.
review, have been highlighted as:
22. Previtali SC, Nodari A, Taveggia C, Pardini C, Dina G, Villa A,
 of special interest Wrabetz L, Quattrini A, Feltri ML: Expression of laminin receptors
 of outstanding interest in schwann cell differentiation: evidence for distinct roles.
J Neurosci 2003, 23:5520-5530.
1. Smith KJ, Blakemore WF, Murray JA, Patterson RC: Internodel
myelin volume and axon surface area. A relationship 23. Yamada H, Denzer A, Hori J, Tanaka H, Fujita S, Fukutaohi H,
determining myelin thickness? J Neurol Sci 1982, 55:231-246. Shimizu T, Ruegg MA, Matsumura K: Dystroglycan is a dual
receptor for agrin and laminin-2 in Schwann cell membrane.
2. Waxman SG: Pathophysiology of demyelinated and J Biol Chem 1996, 271:23418-23423.
remyelinated axons. In: Cook SD. Ed. Handbook of multiple
sclerosis. 2nd ed. New York: Marcel Dekker, 1996: 257-294. 24. Feltri ML, Graus Porta D, Previtali SC, Nodari A, Migliavacca B,
Cassetti A, Littlewood-Evans A, Reichardt LF, Messing A,
3. Webster HD, Martin JR, O’Connell MF: The relationship between Quattrini A et al.: Conditional disruption of beta1 integrin in
interphase Schwann cells and axons before myelination: a Schwann cells impedes interactions with axons. J Cell Biol
quantitative electron microscopic study. Dev Biol 1973, 2002, 156:199-209.
32:401-416.
25. Henry MD, Campbell KP: Dystroglycan inside and out.
4. Salzer JL: Polarized domains of myelinated axons. Curr Opin Cell Biol 1999, 11:602-607.
Neuron 2003, 40:297-318.
26. Rambukkana A, Kunz S, Min J, Campbell KP, Oldstone MB:
5. Poliak S, Peles E: The local differentiation of myelinated axons  Targeting Schwann cells by nonlytic arenaviral infection
at nodes of Ranvier. Nat Rev Neurosci 2003, 4:968-980. selectively inhibits myelination. Proc Natl Acad Sci USA 2003,
6. Scherer SS, Arroyo EJ: Recent progress on the molecular 100:16071-16076.
organization of myelinated axons. J Peripher Nerv Syst 2002, Selective targeting of a-dystroglycan on Schwann cells by LCMV and
7:1-12. Lassa fever virus from the non-lytic Arenavirus family was demonstrated.
Persistent LCMV infection rendered immature Schwann cells incapable of
7. Johnson PC: Peripheral nerve pathology. In text book of forming mature myelin sheath during differentiation, and caused demye-
Neuropathology, 3rd Ed. R.L. Davis, and D.M. Robertson, lination in mature Schwann cells that form myelin sheaths. A correlation
eds. (Baltimore, MD, William and Wilkins) 1997. p. 1233-1323. between the disorganization of the laminin-2 network and downregulation
of a-dystroglycan with myelin defects was demonstrated. Viral manip-
8. Job CK: Nerve damage in leprosy. Int J Lepr Other Mycobact Dis ulation of dystroglycan and laminin-associated functions in Schwann
1989, 57:532-539. cells was proposed for myelination and demyelination. The study also
shows the use of LCMV as a tool to dissect the molecular basis of
9. Waxman SG: Demyelinating diseases-new pathological myelination.
insights, new therapeutic targets. N Engl J Med 1998,
338:323-325. 27. Cao W, Henry MD, Borrow P, Yamada H, Elder JH, Ravkov EV,
Nichol ST, Compans RW, Campbell KP, Oldstone MB:
10. Bjartmar C, Trapp BD: Axonal and neuronal degeneration in Identification of alpha-dystroglycan as a receptor for
multiple sclerosis: mechanisms and functional consequences. lymphocytic choriomeningitis virus and Lassa fever virus.
Curr Opin Neurol 2001, 14:271-278. Science 1998, 282:2079-2081.
11. Hiraga A, Mori M, Ogawara K, Hattori T, Kuwabara S: Differences 28. Masaki T, Matsumura K, Hirata A, Yamada H, Hase A, Arai K,
in patterns of progression in demyelinating and axonal Shimizu T, Yorifuji H, Motoyoshi K, Kamakura K: Expression of
Guillain-Barre syndromes. Neurology 2003, 61:471-474. dystroglycan and the laminin-alpha 2 chain in the rat peripheral
12. Rambukkana A, Zanazzi G, Tapinos N, Salzer JL: Contact nerve during development. Exp Neurol 2002, 174:109-117.
dependent demyelination by Mycobacterium leprae in the 29. Masaki T, Matsumura K, Saito F, Sunada Y, Shimizu T, Yorifuji H,
absence of immune responses. Science 2002, 296:927-931. Motoyoshi K, Kamakura K: Expression of dystroglycan and
13. Ng V, Zanazzi G, Timpl R, Talts JF, Salzer JL, Brennan PJ, laminin-2 in peripheral nerve under axonal degeneration and
Rambukkana A: Role of the cell wall phenolic glycolipid-1 in regeneration. Acta Neuropathol (Berl) 2000, 99:289-295.
the peripheral nerve predilection of Mycobacterium leprae. 30. Hunter SW, Brennan PJ: A novel phenolic glycolipid from
Cell 2000, 103:511-524. Mycobacterium leprae possibly involved in immunogenicity
14. Shimoji Y, Ng V, Matsumura K, Fischetti VA, Rambukkana A: and pathogenicity. J Bacteriol 1981, 147:728-735.
A 21-kDa surface protein of Mycobacterium leprae binds 31. Saito F, Moore SA, Barresi R, Henry MD, Messing A,
peripheral nerve laminin-2 and mediates Schwann cell  Ross-Barta SE, Cohn RD, Williamson RA, Sluka KA, Sherman DL
invasion. Proc Natl Acad Sci USA 1999, 96:9857-9862. et al.: Unique role of dystroglycan in peripheral nerve
15. Rambukkana A, Salzer JL, Yurchenco PD, Tuomanen EI: Neural myelination, nodal structure, and sodium channel stabilization.
targeting of Mycobacterium leprae mediated by the G domain Neuron 2003, 38:747-758.
of the laminin-alpha2 chain. Cell 1997, 88:811-821. A critical role for dystroglycan in myelination and peripheral nerve func-
tions was demonstrated by selective deletion of Schwann cell dystrogly-
16. Rambukkana A, Yamada H, Zanazzi G, Mathus T, Salzer JL, can. Slowed nerve conduction and nodal changes, including reduced
Yurchenco PD, Campbell KP, Fischetti VA: Role of alpha- sodium channel density and disorganized microvilli, were observed in
dystroglycan as a Schwann cell receptor for Mycobacterium mutant mice. Data suggest that dystroglycan is crucial for both myelina-
leprae. Science 1998, 282:2076-2079. tion and normal maintenance of voltage-gated sodium channels at nodes
of Ranvier, possibly by mediating trans interactions between Schwann
17. Talts JF, Andac Z, Gohring W, Brancaccio A, Timpl R: Binding of cell microvilli and the nodal axolemma.
the G domains of laminin alpha1 and alpha2 chains and
perlecan to heparin, sulfatides, alpha-dystroglycan, and 32. Sherman DL, Fabrizi C, Gillespie CS, Brophy PJ: Specific
several extracellular matrix proteins. EMBO J 1999, 18:863-870. disruption of a schwann cell dystrophin-related protein
complex in a demyelinating neuropathy. Neuron 2001,
18. Bunge MB, Bunge RP: Linkage between Schwann cell 30:677-687.
extracellular matrix production and ensheathment function.
Ann N Y Acad Sci 1996, 486:241-247. 33. Gillespie CS, Sherman DL, Fleetwood-Walker SM, Cottrell DF,
Tait S, Garry EM, Wallace VC, Ure J, Griffiths IR, Smith A,
19. Patton BL, Miner JH, Chiu AY, Sanes JR: Distribution and function Brophy PJ: Peripheral demyelination and neuropathic pain
of laminins in the neuromuscular system of developing, adult, behavior in periaxin-deficient mice. Neuron 2000, 26:523-531.
and mutant mice. J Cell Biol 1997, 139:1507-1521.
34. Parkinson DB, Dickinson S, Bhaskaran A, Kinsella MT, Brophy PJ,
20. Uziyel Y, Hall S, Cohen J: Influence of laminin-2 on Schwann Sherman DL, Sharghi-Namini S, Duran Alonso MB, Mirsky R,
cell-axon interactions. Glia 2000, 32:109-121. Jessen KR: Regulation of the myelin gene periaxin provides

www.sciencedirect.com Current Opinion in Immunology 2004, 16:511–518


518 Host–pathogen interactions

evidence for Krox-20-independent my elin-related signalling in 42. Pasare C, Medzhitov R: Toll-like receptors: balancing host
Schwann cells. Mol Cell Neurosci 2003, 23:13-27. resistance with immune tolerance. Curr Opin Immunol 2003,
15:677-682.
35. Scherer SS: Myelination: some receptors required. J Cell Biol
2002, 156:13-15. 43. Krutzik SR, Ochoa MT, Sieling PA, Uematsu S, Ng YW, Legaspi A,
 Liu PT, Cole ST, Godowski PJ, Maeda Y et al.: Activation and
36. Wrabetz L, Feltri ML: Do Schwann cells stop, DR(o)P2, and roll? regulation of Toll-like receptors 2 and 1 in human leprosy.
Neuron 2001, 30:642-644. Nat Med 2003, 9:525-532.
37. Rambukkana A: Molecular basis for the peripheral nerve This study provides evidence that the outcome of the immune response in
predilection of Mycobacterium leprae. Curr Opin Microbiol 2001, leprosy is determined in part by the influence of the local cytokine
4:21-27. environment on the regulated expression and activation of TLR1 and
TLR2. The activation of TLRs at the site of disease was proposed to
38. Mukherjee R, Antia NH: Host-parasite interrelationship between contribute to host defense against microbial pathogens.
M. leprae and Schwann cells in vitro. Int J Lepr Other Mycobact
Dis 1986, 54:632-638. 44. Oliveira RB, Ochoa MT, Sieling PA, Rea TH, Rambukkana A,
Sarno EN, Modlin RL: Expression of Toll-like receptor 2 on
39. Shetty VP, Antia NH: Light and ultrastructural study of sciatic human Schwann cells: a mechanism of nerve damage in
nerve lesions induced using intraneural injection of viable leprosy. Infect Immun 2003, 71:1427-1433.
Mycobacterium leprae in normal and immunosuppressed
Swiss white mice. Int J Lepr Other Mycobact Dis 2002, 70:25-33. 45. Artung HP: Immune-mediated demyelination. Ann Neurol 1993,
33:563-567.
40. Shetty VP, Jacobs JM, Anita NH: The pathology of early leprous
neuropathy. J Neurol Sci 1988, 88:115-135. 46. Spierings E, de Boer T, Wieles B, Adams LB, Marani E,
Ottenhoff TH: Mycobacterium leprae-specific, HLA class
41. Scherer SS, Salzer JL: In: Glial cell development, Jessen KR, II-restricted killing of human Schwann cells by CD4R Th1 cells:
Richardson WD, eds. (Bios Scientific Publishers, Ltd., London, UK a novel immunopathogenic mechanism of nerve damage in
1996; pp. 165-196. leprosy. J Immunol 2001, 166:5883-5888.

Current Opinion in Immunology 2004, 16:511–518 www.sciencedirect.com

You might also like