You are on page 1of 6

Progress in Neurobiology 91 (2010) 102107

Contents lists available at ScienceDirect

Progress in Neurobiology
journal homepage: www.elsevier.com/locate/pneurobio

Usage of signaling in neurodegeneration and regeneration of peripheral nerves by leprosy bacteria


Anura Rambukkana a,b,*
a b

The Rockefeller University, 1230, York Avenue, New York, NY, USA Centers for Neuroregeneration, Regenerative Medicine, and Infectious Diseases, University of Edinburgh, 47, Little France Crescent, Edinburgh EH 16 4BS, Scotland, UK

A R T I C L E I N F O

A B S T R A C T

Article history: Received 15 May 2009 Received in revised form 28 June 2009 Accepted 1 December 2009 Keywords: Signaling Schwann cells Myelination Neurodegeneration De-differentiation Pathogen Neuroregeneration

Multiple signaling pathways play key regulatory roles during the development of peripheral nervous system (PNS) and also in neuroregeneration process following nerve degeneration. Schwann cells, the glial cells of the PNS, by interacting with neuronal (axonal) ligands, mainly neuregulins via receptor tyrosine kinase (RTK) complex, ErbB2/ErbB3, initiate intracellular signaling pathways to drive proliferation and differentiation of Schwann cells, both during development and the process of regeneration and re-myelination after nerve injury. One of the major signaling kinases, extracellular signal-regulated kinase-1/2 (ERK1/2), that is also a downstream signaling pathway of neuregulin-ErbB2/ ErbB3 activation, has been identied as a key regulator of Schwann cell proliferation, differentiation, demyelination and nerve regeneration. Recent studies have provided evidence that the bacterium that causes human leprosy, Mycobacterium leprae that has a unique capacity to invade Schwann cells of the adult PNS, utilizes the neuregulin-ErbB2/ErbB3 associated signaling network to the bacterial advantage. M. leprae directly bind to ErbB2 on myelinated Schwann cells and activate the RTK by a novel route that bypasses the classical neuregulin/growth factor-induced ErbB2-ErbB3 heterodimerization, and subsequently induce downstream the canonical Erk1/2 signaling, leading to myelin breakdown and subsequent axonal damage. This initial injury provides a survival advantage for M. leprae as it induces dedifferentiation and generates myelin-free cells, which are highly susceptible to M. leprae invasion and promote bacterial survival. Once invaded M. leprae activate Erk1/2 via a non-canonical pathway and subsequently increase the cell proliferation and maintain the infected cells in de-differentiated state, thereby preventing remyelination. Therefore, by subverting major RTKs and signaling pathways in adult Schwann cells M. leprae appear to propagate the bacterial niche and maintain survival within the PNS. These studies may also provide new insights into our understanding of signaling mechanisms involve in both neurodegeneration and neuroregeneration. 2009 Elsevier Ltd. All rights reserved.

Contents 1. 2. 3. Introduction: early bacterial life in the peripheral nerves and survival strategies . . . . . . . . . . . . . . . . . . . Schwann cell responses to M. leprae . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Schwann cell signaling and bacterial survival . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1. Role of ErbB2 receptor tyrosine kinase and downstream Erk1/2 signaling in demyelination . . . . . 3.2. Usage of signaling in nerve regeneration as potential mechanisms for M. leprae survival . . . . . . . Targeting cell signaling for prevention of disease progression and as potential therapeutic intervention Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 103 103 104 104 105 105 106 106 106

4. 5.

* Correspondence address: University of Edinburgh, Medical School, Chancellors Building, 47, Little France Crescent, Edinburgh EH16 4BS, Scotland, UK. Fax: +44(0) 131 242 9297. E-mail addresses: rambuka@rockefeller.edu, a.rambuka@ed.ac.uk. 0301-0082/$ see front matter 2009 Elsevier Ltd. All rights reserved. doi:10.1016/j.pneurobio.2009.12.002

A. Rambukkana / Progress in Neurobiology 91 (2010) 102107

103

1. Introduction: early bacterial life in the peripheral nerves and survival strategies One of the classical examples of infectious neurodegenerative diseases of the peripheral nervous system (PNS) is leprosy, which is caused by Mycobacterium leprae and this remains an important global health problem; it is also a leading cause of non-traumatic neuropathies in the world (Job, 1994, 1989; Johnson, 1997; World Health Organization, 2001; Lockwood and Suneetha, 2005; Scollard et al., 2006). M. leprae infection of Schwann cells, the glial cells of the PNS, is the primary cause for the neurological injury, which account for sensory and sensorimotor loss in leprosy patients (Stoner, 1979; Job, 1989). Despite the success of multidrug therapy, which has dramatically reduced the number of leprosy cases worldwide, there has been no reduction in new case detection and obviously the incidence of neurological injuries cannot be reversed by antibiotic treatment alone (World Health Organization, 2001; Lockwood and Suneetha, 2005; Rao, 2005; Scollard et al., 2006). Therefore, it is now recognized that alternative therapeutic strategies, in addition to antibiotics, will be needed to effectively combat the neurological injury, the major complication that continues to be a problem in endemic areas. Understanding the molecular basis by which M. leprae cause functional defects and induce neurodegeneration is of signicant medical importance, not only for preventing the disease progression at the early stage, but also for the insight such knowledge provides into basic biology of neurodegenerative diseases in general. In the PNS, Schwann cells not only serve as a natural host but also a safe haven for the multiplication of M. leprae since the bloodnerve barrier protects the organism from the host immune responses (Lumsden, 1959; Stoner, 1979; Johnson, 1997). Therefore, Schwann cell resident M. leprae serve as a primary source of infection, which not only causes nerve injury, but are also responsible for the continuous leakage of M. leprae and ultimately facilitate dissemination of infection to other non-neural tissues including smooth and skeletal muscles often seen in various stages in leprosy patients (Job, 1994; Johnson, 1997; Scollard et al., 2006). Because M. leprae is an obligate intracellular pathogen with the longest doubling time among bacteria and limited number of functional genes in its genome (Cole et al., 2001), the establishment of productive infection within Schwann cells is the key for bacterial survival in the PNS. In human infection it is likely that M. leprae reside and replicate within Schwann cells for a long period before the immune cells actively come into play and initiate inammatory responses, which eventually manifest clinically as sensory or sensorimotor loss (Job, 1989; Johnson, 1997). Prior to this stage, from initial infection to the rst symptoms of nerve involvement, we know nothing about what is happening in early human infection. This initial phase is a completely a black box, but is critical for the propagation of M. leprae and the establishment of bacterial niche within the PNS and subsequent nerve damage and disease progression. Recent studies with in vivo grown M. leprae (Truman and Krahenbuhl, 2001) have contributed to our understanding of how M. leprae subvert the Schwann cell functions and the potential early life cycle of this unique bacterium within the PNS (Rambukkana et al., 1997, 1998, 2002; Ng et al., 2000; Rambukkana, 2004; Tapinos and Rambukkana, 2005; Tapinos et al., 2006). Schwann cells exist in the adult PNS as two phenotypes, myelinating Schwann cells that form one-to-one relationships with large caliber axons and non-myelinating Schwann cells that ensheath multiple small axons (Webster et al., 1973). During development both Schwann cell phenotypes are derived from the neural crest (Jessen and Mirsky, 2005). Myelinated nerve bers enable rapid saltatory impulse propagation due to the larger

caliber axon and the myelin sheath, whereas the non-myelinated bers mostly comprised of smaller sensory axons, provide slow nerve conduction (Sherman and Brophy, 2005; Nave and Salzer, 2006). Clinically, early leprosy is characterized by sensory neuropathies due to injuries to sensory axons (Job, 1994, 1989; Johnson, 1997). Studies with myelinating Schwann cell-neuron cocultures have shown that myelinated and non-myelinated Schwann cells exhibit distinct functional responses to M. leprae infection (Rambukkana et al., 2002). Although M. leprae binds to both myelinated and non-myelinated Schwann cell-axon units, it is the non-myelinated Schwann cells that are highly susceptible to M. leprae invasion and preferentially harbor M. leprae in peripheral nerves (Johnson, 1997; Shetty et al., 1980; Shetty and Antia, 1988; Rambukkana et al., 2002). On the other hand, myelinated Schwann cells are mostly resistant to M. leprae invasion, but attachment of the bacteria induces demyelination and axonal damage (Rambukkana et al., 2002; Fig. 1). The resistance of myelinated Schwann cells to M. leprae invasion may be due to the compact myelin sheath that physically occupies almost the entire cytoplasm of the cells. Indeed, the myelin sheath is a clear obstacle, both physically and physiologically for the long-term intracellular residence, survival and replication of M. leprae. Interestingly, early histopathological studies of nerve biopsies from lepromatous leprosy patients have also shown that intracellular M. leprae are frequently seen in non-myelinated Schwann cells in the early stage, but bacilli are rarely seen in myelinating Schwann cells (4%) even in advanced lepromatous leprosy patients with high bacterial load (Shetty et al., 1980; Shetty and Antia, 1988). These data reinforce in vitro nding that non-myelinated or myelin-free Schwann cells are needed for effective invasion and intracellular survival. This earliest phase of infection is critical for the establishment of bacterial niche and the outcome of disease progression and nerve damage. Although we know nothing about this earliest phase in patients it is quite possible that demyelination could occur in very early stage, but there is no way of knowing it due to lack of diagnosis. Further, the non-immune mediated myelin damage could rapidly be repaired as remyelination is known to occur as a response to peripheral nerve injury (Fawcett and Keynes, 1990; Stoll et al., 2002). 2. Schwann cell responses to M. leprae Schwann cells are notoriously sensitive to environmental changes, that can affect cellular behavior and functions. These changes inuence cells decision to remain or become quiescent, to differentiate, to undergo programmed cell death or to proliferate and de-differentiate. M. leprae appear to have evolved to induce cell proliferation and de-differentiation after they induce demyelination and invade non-myelinated Schwann cells to generate an intracellular niche for long-term bacterial survival and to compensate for its limited number of functional genes (Rambukkana et al., 2002; Johnson, 1997; Shetty et al., 1980; Shetty and Antia, 1988; Fig. 1). In adult non-myelinated Schwann cells M. leprae can be seen in large numbers in lepromatous leprosy patients (Lumsden, 1959; Johnson, 1997; Shetty et al., 1980; Shetty and Antia, 1988). These ndings suggest that M. leprae multiply as long as Schwann cells, which lack of anti-microbicidal machinery, can tolerate the bacterial load and then release and infect more Schwann cells (Rambukkana et al., 2002, 2004; Fig. 1). The strictly obligate intracellular parasitic nature requires M. leprae to rapidly invade its susceptible target in order to survive. However, as the infection progresses and bacteria undergo unrestrained multiplication, the availability of adult non-myelinating Schwann cells becomes a limiting factor. To avoid such a situation, M. leprae have evolved to activate the quiescent non-myelinated Schwann cells to re-enter the cell-division cycle and proliferate, thereby generating

104

A. Rambukkana / Progress in Neurobiology 91 (2010) 102107

Fig. 1. A model for intracellular life of M. leprae in the PNS and the role of signaling pathways. Upon infection, M. leprae attach to both myelinated and non-myelinated Schwann cells, the two functional units of the PNS, but invade preferentially non-myelinated phenotypes where they multiply and then release and re-infect non-myelinated Schwann cells and induce cell proliferation. To continue this intracellular life cycle within its preferred niche and to establish productive infection, M. leprae have adapted to generate myelin-free Schwann cell pool (i) by causing nerve injury, demyelination and axonal damage, which is important to repair the damaged nerve (nerve regeneration) and (ii) directly by proliferating non-myelinated Schwann cells in response to intracellular M. leprae. Infected myelin-free de-differentiated Schwann cells fail to undergo proper re-differentiation program as intracellular M. leprae inhibit this process, thereby maintaining the myelin-free Schwann cell pool. These distinct cellular responses by extracellular and intracellular M. leprae eventually generate myelin-free de-differentiated Schwann cells, which are not only highly susceptible for M. leprae invasion but also provide a comfortable niche for bacterial multiplication and establishment within the PNS. Multiple signaling pathways, predominantly the Erk1/2 signaling via canonical and non-canonical pathways, downstream of direct activation of ErbB2 and neuregulin-ErbB3/ErbB2 network are involved in different stages of the early infectious process.

more myelin-free cells (Fig. 1). On the other hand, extracellular M. leprae induce demyelination and subsequent axonal damage and this condition is somewhat similar to peripheral nerve injury in vivo where Schwann cells rapidly proliferate and enclose the injured axons, an important event for the promotion of nerve regeneration (Scherer and Salzer, 1996; Fawcett and Keynes, 1990; Stoll et al., 2002). Strikingly, Schwann cells proliferate in M. lepraetreated nerve tissue cultures with substantial demyelination and axonal damage (Rambukkana et al., 2002). Such a microenvironment created by M. leprae infection may also contribute to Schwann cell proliferation. Both of these distinct cellular responses to extracellular and intracellular M. leprae appear to generate myelin-free Schwann cells that are highly susceptible for M. leprae invasion (Rambukkana et al., 2002; Tapinos and Rambukkana, 2005; Tapinos et al., 2006) (Fig. 1). Furthermore, the non-toxic nature of M. leprae is an additional advantage that ensures Schwann cell survival during long-term incubation without inducing apoptosis or toxicity (Tapinos and Rambukkana, 2005). Therefore, it is of interest and importance to understand how intracellular M. leprae regulates Schwann cell functions and how these functions inuence the maintenance of long-term M. lepraeSchwann cell relationship, that subsequently leads to impairment of normal neurological functions or regenerate the damaged nerves for the benet of bacterial survival. 3. Schwann cell signaling and bacterial survival Signaling pathways play key regulatory role during the developmental of Schwann cells and neuroregeneration process following nerve injury (Guertin et al., 2005; Grove et al., 2007; Mirsky et al., 2008; Jessen and Mirsky, 2008; Nave and Trapp, 2008).

Schwann cells by interacting with neuronal (axonal) ligands, mainly neuregulins initiate intracellular signaling pathways to drive proliferation and subsequent differentiation/myelination of Schwann cells (Garratt et al., 2000; Riethmacher et al., 1997; Lemke, 2006; Nave and Salzer, 2006; Harrisingh et al., 2004; Jessen and Mirsky, 2008). Recent studies now suggest that the Schwann cell differentiation process, particularly myelination can be viewed as a balance between positive and negative regulators by both signaling pathways and transcription factors (Parkinson et al., 2008; Jessen and Mirsky, 2008). In vivo, negative regulators can be induced by nerve injury or removal of Schwann cells from axonal contacts (Jessen and Mirsky, 2008). When M. leprae invade adult peripheral nerves it causes a similar situation and is likely to deregulate the existing balance between negative and positive regulators of myelination. Since myelination is not a favorable condition for intracellular survival of M. leprae the positive balance that usually dominate in normal nerves is likely to shift to negative after infection and subsequent demyelination. One of the major signaling kinases that is upregulated after nerve injury is extracellular signalregulated kinase-1/2 (ERK1/2) (Harrisingh et al., 2004). Recent studies have identied how extracellular M. leprae induce demyelination by activating demyelinating Erk1/2 signaling in myelinated Schwann cells (Tapinos et al., 2006). 3.1. Role of ErbB2 receptor tyrosine kinase and downstream Erk1/2 signaling in demyelination In the PNS, activation of signaling can be mediated by growth factor or ligand-dependent phosphorylation of receptor tyrosine kinase (RTK) (Garratt et al., 2000; Riethmacher et al., 1997; Lemke, 2006; Guertin et al., 2005). Induction of demyelination in

A. Rambukkana / Progress in Neurobiology 91 (2010) 102107

105

peripheral nerves has been shown in response to exogenous axonal mitogens such as neuregulins (Zanazzi et al., 2001; Harrisingh et al., 2004). ErbB family receptor ErbB2 is one of the major RTKs expressed on Schwann cells and plays an important role in glial cell functions (Riethmacher et al., 1997; Garratt et al., 2000; Guertin et al., 2005; Lemke, 2006). Among the members of ErbB family, ErbB2 is considered as a ligandless receptor that transduces strong signals by avid dimerization with other ErbB members (Yarden and Sliwkowski, 2001; Hynes and Lane, 2005). Overexpression of ErbB2 is linked to the development of breast and ovarian cancers, and anti-ErbB2 blocking antibody Herceptin and pharmacological inhibitors such as PKI-166 that specically disrupt ErbB2 kinase activity have shown promising results in managing tumor progression in patients and animal models (Yarden and Sliwkowski, 2001; Hynes and Lane, 2005). In Schwann cells, ErbB2 is known to process the signaling by dimerization with ErbB3 after neuregulin binds to ErbB3, which does not have a kinase domain (Garratt et al., 2000). Strikingly, M. leprae directly bind to and activate ErbB2 without ErbB3 heterodimerization (Tapinos et al., 2006). This binding is sufcient to induce early demyelination following activation of ErbB2 by a novel route that bypasses the classical neuregulin/growth factor-induced ErbB2-ErbB3 heterodimerization and subsequently induces downstream MEK-dependent Erk1/2 signaling pathway, leading to myelin breakdown (Garratt et al., 2000; Tapinos et al., 2006). Interestingly, Erk1/2 is known to be a key regulator of Schwann cell differentiation, and activation of this pathway by neuregulin also induces cell proliferation and demyelination (Zanazzi et al., 2001; Harrisingh et al., 2004). Erk1/2 is also strongly activated in Schwann cells during nerve regeneration following nerve injury in vivo (Harrisingh et al., 2004; Noon and Lloyd, 2005). One of the signaling pathways that is strongly activated in Schwann cells during nerve regeneration process following nerve injury is the Erk1/2 (Harrisingh et al., 2004). Therefore, it is likely that M. leprae take advantage of the existing Erk1/2 signaling that is involved in both nerve degeneration and regeneration. 3.2. Usage of signaling in nerve regeneration as potential mechanisms for M. leprae survival Unlike central nervous system (CNS) neurons, the neurons in the PNS can regenerate when damaged; this differential response to injury resides largely within the proliferative and other properties of Schwann cells (Scherer and Salzer, 1996; Fawcett and Keynes, 1990; Stoll et al., 2002; Noon and Lloyd, 2005). Following nerve injury, adult Schwann cells undergo proliferation and one of the key regulators of the cell cycle, cyclin D1 plays a role in this re-entry into the cell-division cycle. These nerve injury responses lead to the generation of new de-differentiated Schwann cells, which facilitate the remyelination and regeneration of the damaged axons by initiating Schwann cell differentiation program (Kim et al., 2000; Scherer and Salzer, 1996; Fawcett and Keynes, 1990; Stoll et al., 2002). M. leprae appear to mimic the events following nerve injury since intracellular M. leprae induces the accumulation of cyclin D1 in infected Schwann cells and subsequently increases the cell division (Tapinos and Rambukkana, 2005). When newly generated Schwann cells or similar de-dedifferentiated Schwann cells are infected by M. leprae they fail to ensheath properly and myelinate since intracellular bacteria prevent these cells to re-differentiate and form the compact myelin sheath (Fig. 1; unpublished data). A similar situation could also occur when M. leprae infect adult peripheral nerves in humans as de-differentiated Schwann cells generated following demyelination and axonal damage could be reinfected by M. leprae and subsequently inhibit their remyelination. These ndings suggest that intracellular M. leprae turn off the Schwann cell differentiation program and maintain the infected cells in an

undifferentiated stage. Erk1/2 signaling is strongly activated following nerve injury and de-differentiation process in vivo (Harrisingh et al., 2004). Similarly, intracellular M. leprae also activate Erk1/2 signaling in de-differentiated human Schwann cells by a novel route using non-canonical MEK-independent and protein kinase C-e (PKC-e) and lymphocyte kinase (Lck) dependent pathway (Tapinos and Rambukkana, 2005). Therefore, it is possible that continuing activation of non-canonical ERK1/2 signaling pathway in infected Schwann cells maintain cells in de-differentiated state and thus prevent remyelination. In addition, our recent studies suggest that intracellular M. leprae further exploit the existing neuregulinErbB2/ErbB3 signaling network by inhibiting ErbB3 function in Schwann cell differentiation, particularly the nuclear translocation of ErbB3, which appears to play a role in myelin gene expression and myelination (unpublished data). Together, these ndings suggest that M. leprae has evolved to utilize existing neuregulin-ErbB3/ErbB2 signaling network for modulating Schwann cell functions. This bacterial strategy using signaling mediated demyelination and cell proliferation may provide a successful survival mechanism by facilitating the propagation of de-differentiated cells suitable for the life style of leprosy bacteria. 4. Targeting cell signaling for prevention of disease progression and as potential therapeutic intervention Because the humanized anti-ErbB2 Herceptin has shown to block the binding of M. leprae to ErbB2 on human Schwann cells and inhibits the demyelinating signaling it is possible that an intervention at this stage by therapeutic means could prevent early demyelination (Tapinos et al., 2006). The ndings that M. leprae bind to and activate ErbB2 suggest that M. leprae carry a ligand(s) for ErbB2 receptor on Schwann cells and such ligand(s) could serve as a trigger for early nerve damage during infection, and may also serve as a basis for developing therapeutics for leprosy-induced nerve damage. For example, an antagonist(s) that nullies the effect of bacterial ErbB2 ligand(s) could prevent demyelination signaling induced by M. leprae via ErbB2 activation. Inhibition of ErbB2 activation by pharmacological agents can inhibit the demyelination at two critical points, one at the activation of ErbB2 level and another at the downstream signaling at MEK activation using wellcharacterized inhibitors (Fig. 2) that are currently used in cancer therapy or clinical trials of cancer patients (Yarden and Sliwkowski, 2001; Hynes and Lane, 2005; Franklin and Zhao, 2006). The direct application of the nding of ErbB2 and MEK/Erk1/2 involvement in M. leprae-induced demyelination has the potential of therapeutic use of specic small molecule kinase inhibitors to these signaling targets in the management of leprosy nerve damage (Fig. 2) and perhaps for other neurodegenerative diseases (Franklin and Zhao, 2006). Because ErbB2 activation and MEK/Erk1/2 signaling are well documented in a variety of cancers, several promising small molecule drugs are now available and that are in various phases of clinical trials in cancer patients (Hynes and Lane, 2005). On the other hand, in the course of nerve injury Schwann cells begin to actively divide which in turn, facilitates the regeneration of the damaged nerves (Fawcett and Keynes, 1990; Stoll et al., 2002). M. leprae use early demyelination as a survival strategy for generating more myelin-free Schwann cells to expand the intracellular bacterial niche for long-term survival and thus prevention of this response would likely facilitate the control of the infection (Fig. 1). These early events following M. leprae infection, particularly the occurrence of demyelination and axonal injury and cellular responses to such nerve injury provides an opportunity to identify marker molecules that are indicative of nerve injury (Fig. 2). Therefore, the changes in neural microenvironment as a consequence of nerve injury responses to early

106

A. Rambukkana / Progress in Neurobiology 91 (2010) 102107

Fig. 2. Targeting cell signaling for potential therapeutic intervention. Understanding M. leprae strategies will provide the targets for new therapeutics to combat the neurological injury and disease progression. Several intervention stages can be proposed for therapeutic and/or diagnostic approaches using the current knowledge of how M. leprae subvert RTK and Erk1/2 signaling pathways in the PNS. (A) Early M. leprae-Schwann cell interaction: the ndings that M. leprae bind to and activate ErbB2 on myelinated Schwann cells suggest that M. leprae carry a ligand(s) for ErbB2 receptor and such ligand(s) could serve as a trigger for early nerve damage. An antagonist(s) that nullies the effect of bacterial ErbB2 ligand(s) could be an effective reagent for preventing demyelinating signaling. (B) ErbB2 and Erk1/2 activation: Humanized anti-ErbB2 Herceptin blocks the binding of M. leprae to ErbB2 on human Schwann cells and inhibits the demyelinating signaling. Intervention at this stage by therapeutic means could prevent early demyelination. Inhibition of ErbB2 activation by pharmacological agents could inhibit the demyelination at two critical points, one at the activation of ErbB2 level and another at the downstream signaling at MEK activation using well-characterized inhibitors that are currently used in cancer therapy or clinical trials of cancer patients. (C) Nerve injury response: M. leprae-induced nerve injury should facilitate the regeneration of the damaged nerves. Cellular responses to such nerve injury provide an opportunity to identify marker molecules that are indicative of nerve injury, and thus provide information for developing potential diagnostics or therapeutic interventions.

demyelination may provide information for developing diagnostics or therapeutic interventions before the disease is aggravated by immune attack and cripples the patients. 5. Conclusion We are beginning to understand the mechanisms by which M. leprae exploit the critical Schwann cell functions for the long-term bacterial survival, which enable this strictly obligate intracellular bacterium to establish productive infection within the PNS and subsequently induce neurodegeneration and disseminate the infection into other tissues. It is hoped that targeting M. leprae strategies at this very early stage will provide the rationale to develop diagnostics and new therapeutics to combat the neurological injury and disease progression. It is important to further understand how M. leprae establish the productive infection within the PNS at the earliest stage, that eventually lead to neurodegeneration, dissemination and transmission of infection. Our current knowledge suggests that M. leprae utilize the major neuregulinErbB2/ErbB3 signaling network to manipulate critical Schwann cell functions, myelination, demyelination, de-differentiation and proliferation, thereby maintain long-term bacterial survival within the PNS. These studies may provide rational to develop novel therapeutics to prevent the nerve damage during infection and the disease progression. Importantly, detail analyses of the

mechanisms by which M. leprae induce demyelination, cell proliferation and inhibit the myelination program may provide valuable information for our understanding of early stage of neurodegeneration and nervous system repair processes, which we know very little about. Acknowledgements I thank members of my laboratory, both past and present members, particularly Nikos Tapinos, Toshihiro Masaki, Ryan Raaum, Joesph Dybass, Hanpeng Xu and Jinrong Qu for contributing to the published and unpublished studies from which this review was made possible. The research in the authors laboratory is supported by R01 grants from NINDS and NIAID, Maltalepra Foundation (France) and US National Multiple Sclerosis. References
Cole, S.T., Eiglmeier, K., Parkhill, J., James, K.D., Thomson, N.R., Wheeler, P.R., Honore, N., Garnier, T., Churcher, C., Harris, D., Mungall, K., Basham, D., Brown, D., Chillingworth, T., Connor, R., Davies, R.M., Devlin, K., Duthoy, S., Feltwell, T., Fraser, A., Hamlin, N., Holroyd, S., Hornsby, T., Jagels, K., Lacroix, C., Maclean, J., Moule, S., Murphy, L., Oliver, K., Quail, M.A., Rajandream, M.A., Rutherford, K.M., Rutter, S., Seeger, K., Simon, S., Simmonds, M., Skelton, J., Squares, R., Squares, S., Stevens, K., Taylor, K., Whitehead, S., Woodward, J.R., Barrell, B.G., 2001. Massive gene decay in the leprosy bacillus. Nature 409, 10071011.

A. Rambukkana / Progress in Neurobiology 91 (2010) 102107 Fawcett, J.W., Keynes, R.J., 1990. Peripheral nerve regeneration. Annu. Rev. Neurosci. 13, 4360. Franklin, R.J.M., Zhao, C., 2006. Tyrosine kinases: maiming myelin in leprosy. Nat. Med. 12, 889890. Garratt, A.N., Britsch, S., Birchmeier, C., 2000. Neuregulin, a factor with many functions in the life of a Schwann cell. Bioessays 22, 987996. Grove, M., Komiyama, N.H., Nave, K.A., Grant, S.G., Sherman, D.L., Brophy, P.J., 2007. FAK is required for axonal sorting by Schwann cells. J. Cell Biol. 176, 277282. Guertin, A.D., Zhang, D.P., Mak, K.S., Alberta, J.A., Kim, H.A., 2005. Microanatomy of axonal/glial signaling during Wallerian degeneration. J. Neurosci. 25, 34783487. Harrisingh, M.C., Perez-Nadales, E., Parkinson, D.B., Malcolm, D.S., Mudge, A.W., Lloyd, A.C., 2004. The Ras/Raf/ERK signaling pathway drives Schwann cell dedifferentiation. EMBO J. 23, 30613071. Hynes, N.E., Lane, H.A., 2005. ERBB receptors and cancer: the complexity of targeted inhibitors. Nat. Rev. Cancer 5, 341354. Jessen, K.R., Mirsky, R., 2008. Negative regulation of myelination: relevance for development, injury, and demyelinating disease. Glia 56, 15521565. Jessen, K.R., Mirsky, R., 2005. The origin and development of glial cells in peripheral nerves. Nat. Rev. Neurosci. 671682. Job, C.K., 1994. Pathology of leprosy. In: Hastings, R.S., Opromolla, D.V.A. (Eds.), Leprosy. 2nd ed. Churchill Livingstone, Philadelphia, pp. 190233. Job, C.K., 1989. Nerve damage in leprosy. Int. J. Lepr. Other Mycobact. Dis. 57, 532539. Johnson, P.C., 1997. Peripheral nerve pathology. In: Davis, R.L., Robertson, D.M. (Eds.), Text Book of Neuropathology. 3rd ed. William and Wilkins, Baltimore, MD, pp. 12331323. Kim, H.A., Pomeroy, S.L., Whoriskey, W., Pawlitzky, I., Benowitz, L.I., Sicinski, P., Stiles, C.D., Roberts, T.M., 2000. A developmentally regulated switch directs regenerative growth of Schwann cells through cyclin D1. Neuron 26, 405416. Lemke, G., 2006. Neuregulin-1 and myelination. Sci. STKE, pe11. Lockwood, D.N., Suneetha, S., 2005. Leprosy: too complex a disease for a simple elimination paradigm. Bull. Worlds Health Organ. 83, 230235. Lumsden, C., 1959. Leprosy and the Schwann cell in vitro and in vivo. In: Cochrane, R. (Ed.), Leprosy in Theory and Practice. John Wright and Sons, Ltd., Bristol, UK, pp. 221250. Mirsky, R., Woodhoo, A., Parkinson, D.B., Arthur-Farraj, P., Bhaskaran, A., Jessen, K.R., 2008. Novel signals controlling embryonic Schwann cell development, myelination and dedifferentiation. J. Peripher. Nerv. Syst. 13, 122135. Nave, K.A., Salzer, J.L., 2006. Axonal regulation of myelination by neuregulin 1. Curr. Opin. Neurobiol. 16, 492500. Nave, K.A., Trapp, B.D., 2008. Axon-Glial Signaling and the support of axon function. Annu. Rev. Neurosci. 31, 535561. Ng, V., Zanazzi, G., Salzer, J.L., Timpl, R., Talts, J.F., Brennan, P., Rambukkana, A., 2000. Role of the cell wall Phenolic Glycolipid-1 in the peripheral nerve predilection of Mycobacterium leprae. Cell 103, 511524. Noon, L.A., Lloyd, A.C., 2005. Hijacking the ERK signaling pathway: Mycobacterium leprae shuns MEK to drive the proliferation of infected Schwann cells. Sci. STKE 309, pe52. Parkinson, D.B., Bhaskaran, A., Arthur-Farraj, P., Noon, L.A., Woodhoo, A., Lloyd, A.C., Feltri, M.L., Wrabetz, L., Behrens, A., Mirsky, R., Jessen, K.R., 2008. c-Jun is a negative regulator of myelination. J. Cell. Biol. 181, 625637.

107

Rambukkana, A., Salzer, J.L., Yurchenco, P.D., Tuomanen, E.I., 1997. Neural targeting Mycobacterium leprae mediated by the G domain of the laminin alpha2 chain. Cell 88, 811821. Rambukkana, A., Yamada, H., Sanazzi, G., Salzer, J.L., Yurchenco, P.D., Campbell, K.P., 1998. Role of alpha-dystroglycan as a Schwann cell receptor for Mycobacterium leprae. Science 282, 20762079. Rambukkana, A., Zanazzi, G., Tapinos, N., Salzer, J.L., 2002. Contact-dependent demyelination by Mycobacterium leprae in the absence of immune responses. Science 296, 927931. Rambukkana, A., 2004. Mycobacterium leprae-induced demyelination: a model for early nerve degeneration. Curr. Opin. Immunol. 16, 511518. Riethmacher, D., Sonnenberg-Riethmacher, E., Brinkmann, V., Yamaai, T., Lewin, G.R., Birchmeier, C., 1997. Severe neuropathies in mice with targeted mutations in the ErbB3 receptor. Nature 725730. Rao, N., 2005. Leprosy program in India at the crossroads. Int. J. Lepr. 73, 211215. Scherer, S.S., Salzer, J.L., 1996. In: Jessen, K.R., Richardson, W.D. (Eds.), Glial Cell Development. Bios Scientic Publishers, Ltd., London, UK, pp. 165196. Sherman, D.L., Brophy, P.J., 2005. Mechanisms of ensheathment and myelin growth. Nat. Rev. Neurosci. 683690. Shetty, V.P., Antia, N.H., Jacobs, J.M., 1988. The pathology of early leprous neuropathy. J. Neurol. Sci. 88, 115131. Shetty, V.P., Mehta, L.N., Irani, P.F., Antia, N.H., 1980. Study of the evolution of nerve damage in leprosy. Part I. Lesions of the index branch of the radial cutaneous nerve in early leprosy. Lepr. India 52, 518. Scollard, D.M., Adams, L.B., Gillis, T.P., Krahenbuhl, J.L., Truman, R.W., Williams, D.L., 2006. The continuing challenges of leprosy. Clin. Microbiol. Rev. 19, 338 381. Stoll, G., Jander, S., Myers, R.R., 2002. Degeneration and regeneration of the peripheral nervous system: from Augustus Wallers observations to neuroinammation. J. Peripher. Nerv. Syst. 7, 1327. Stoner, G.L., 1979. Importance of the neural predilection of Mycobacterium leprae in leprosy. Lancet 2 (8150), 994996. Tapinos, N., Ohnishi, M., Rambukkana, A., 2006. ErbB2 receptor tyrosine kinase mediates early demyelination induced by leprosy bacilli. Nat. Med. 12, 961 966. Tapinos, N., Rambukkana, A., 2005. Insight into regulation of human Schwann cell proliferation by Erk1/2 via MEK-independent and p56Lck-dependent pathway from leprosy bacilli. Proc. Natl. Acad. Sci. U.S.A. 102, 91889193. Truman, R.W., Krahenbuhl, J.L., 2001. Viable Mycobacterium leprae as a research reagent. Int. J. Lepr. 69, 112. Webster, H.D., Martin, R., OConnell, M.F., 1973. The relationships between interphase Schwann cells and axons before myelination: a quantitative electron microscopic study. Dev. Biol. 32, 401416. World Health Organization: Leprosy Fact Sheet, 2001, http://www.who.int/inf-fs/ en/fact101.htmll. Yarden, Y., Sliwkowski, M.X., 2001. Untangling the ErbB signalling network. Nat. Rev. Mol. Cell. Biol. 2, 127137. Zanazzi, G., Einheber, S., Westreich, R., Hannocks, M.J., Bedekk-Hogan, D., Salzer, J., 2001. Glial growth factor/neuregulin inhibits Schwann cell myelination and induces demyelination. J. Cell. Biol. 152, 12891299.

You might also like