Frontier Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Neurol. Dec 28, 2013; 3(4): 87-96
Published online Dec 28, 2013. doi: 10.5316/wjn.v3.i4.87
Cannabinoids: Do they have the potential to treat the symptoms of multiple sclerosis?
Zubair Ahmed, Neurotrauma and Neurodegeneration Section, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, United Kingdom
Author contributions: Ahmed Z participated in research design, conducted experiments, performed data analysis and wrote the manuscript.
Supported by The University of Birmingham
Correspondence to: Dr. Zubair Ahmed, Neurotrauma and Neurodegeneration Section, School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, Institute of Biomedical Research (West), Edgbaston, Birmingham B15 2TT, United Kingdom. z.ahmed.1@bham.ac.uk
Telephone: +44-121-4148859 Fax: +44-121-4148867
Received: May 8, 2013
Revised: September 24, 2013
Accepted: October 16, 2013
Published online: December 28, 2013

Abstract

This article reviews the role of cannabinoids in inhibiting neurodegeneration in models of multiple sclerosis (MS). MS is a chronic, debilitating disease of the central nervous system (CNS), induced by autoimmunity-driven inflammation that leads to demyelination and thus disconnection of the normal transmission of nerve impulses. Despite the use of an array of immune modulating drugs that restore blood brain barrier function, disability continues in patients concomitant with the loss of axons in the spinal cord. MS patients therefore suffer neuropathic pain, spasticity and tremor. Anecdotal evidence suggests that MS patients using cannabis, though illegal, achieve symptomatic relief from neuropathic pain and spasticity associated with MS. The discovery of the endogenous cannabinoid (endocannabinoid) system that naturally exists in the body and which responds to cannabinoids to exert their effects has aided research into the therapeutic utility of cannabinoids. The endocannabinoid system consists of two G-protein coupled receptors cannabinoid receptor type-1 (CB1) and CB2. CB1 is mainly expressed in the CNS and CB2 is predominantly found in leukocytes, while an increasing number of potential ligands and endocannabinoid degradation molecules are being isolated. Several studies have highlighted the involvement of this system in regulating neurotransmission and its ability to prevent excessive neurotransmitter release, consistent with a capacity to provide symptomatic relief. In summary, antagonism of the CB1 receptor pathway contributes to neuronal damage in chronic relapsing experimental allergic encephalomyelitis (EAE) and suppresses tremor and spasticity. The addition of exogenous CB1 agonists derived from cannabis also afforded significant neuroprotection from the consequences of inflammatory CNS disease in EAE and experimental allergic uveitis models. Although clear neuroprotective benefits of cannabinoids have been demonstrated, the unwanted psychotropic effects need to be addressed. However, manipulating the endogenous cannabinoid system may be one way of eliciting beneficial effects without some or all of the unwanted side effects.

Key Words: Multiple sclerosis, Axonal damage, Neurodegeneration, Neuroprotection

Core tip: Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system and causes disability, neuropathic pain, spasticity and tremor in affected patients. Although illegal, users of cannabis report relief from pain and spasticity, probably due to the endogenous cannabinoid system that exists. Cannabinoid receptor type-1 (CB1)-deficient mice accrue greater levels of neurodegeneration and poorly tolerate inflammatory and excitotoxic insults after immune attack in a model of MS, experimental allergic encephalomyelitis. Treatment of animals affected by experimental allergic uveitis (EAU) with CB1 agonists also provided significant neuroprotection from the consequences of EAU, suggesting that cannabinoids may slow down neurodegeneration in MS.



BIOGRAPHY

Dr Zubair Ahmed received his PhD from University College London, London, United Kingdom. He completed his first postdoctoral training period at the Institute of Neurology in London before moving to the University of Birmingham to continue his second postdoctoral training period. In 2007, he was awarded an Research Councils United Kingdom Academic Fellowship, the remit of which was to develop an independently funded research group before being promoted to Lecturer in 2011. He is currently a Senior Lecturer in Neuroscience. His research interests covers numerous aspects of the molecular biology of central nervous system (CNS) axon regeneration and degeneration and his most notable contribution is to the understanding of the role of caspase-2 in apoptosis of retinal and dorsal root ganglion neurons, the contribution of an endogenous mechanism for receptor shedding, the identification of growth factors capable of promoting retinal ganglion cell survival and axon regeneration, identification of alternative neuronal receptors capable of interacting with key molecules in blocking CNS axon regeneration and the observations that cannabinoids inhibit neurodegeneration in models of multiple sclerosis (MS). His curriculum vitae lists over 50 peer-reviewed publications, 2 book chapters and numerous presentations at national and international meetings.

INTRODUCTION

MS is an inflammatory demyelinating disease of the CNS and results in disruption to the normal transmission of nerve impulses due to lesions in the CNS[1,2]. Despite immune modulating drugs that reduce blood brain barrier dysfunction, disability often continues in patients and suggests that neurodegenerative changes are key to the progression of disease[3-5]. MS patients thus display spasticity, neuropathic pain associated with neuroinflammation, excitotoxicity and chronic neurodegeneration. It is also established that axonal/neuronal loss, which occurs early in the disease process, is an important contributor of permanent disability and is often associated with active inflammation. Disability only becomes evident when normal compensatory mechanisms are exhausted, while demyelinated axons are left vulnerable to further damage by electrical activity[6,7]. Therefore identifying neuroprotective strategies are a key goal in the fight against MS.

Although the Cannabis sativa plant has been used for centuries as a medicinal preparation to relieve the symptoms of inflammatory and neuropathic disorders, its use is illegal[8]. However, anecdotal accounts from MS patients indicated that cannabis might offer symptomatic relief of pain and spasticity associated with MS[9]. Plant-derived “cannabinoids” have provided important insights into the biology of cannabis and have led to a multitude of clinical trials using cannabinoids to control pain and spasticity in MS patients[10]. The main active ingredient of Cannabis sativa was defined in 1964 as (-)-trans-delta-9-tetrahydrocannabinol [Δ9-THC or dronabinol (international non-proprietary name)][11]. Δ9-THC is not only responsible for the majority of the pharmacological actions of cannabis but also its psychoactive effects. Numerous other cannabinoids and phytochemicals also exist in the cannabis plant including cannabidiol (CBD) which is non-psychoactive and is not a cannabinoid receptor agonist[12].

CLINICAL TRIALS OF CANNABIS IN MS

Although many patients self-medicated with cannabis, there were few clinical studies that demonstrated reliable clinical evidence of the benefits of cannabis use in MS. Some of the first few studies on the effect of Δ9-THC were not encouraging since small sample sizes were used and Δ9-THC had no effect on objective measures, despite patients reporting subjective improvements in the symptoms of MS[13]. For example, the first systematic, placebo-controlled trial with Δ9-THC and a Cannabis sativa plant extract, given to 16 MS patients with severe spasticity, showed no effect of the cannabinoids on spasticity[14]. A much larger trial involving 660 MS patients receiving Δ9-THC or natural Cannabis oil or a placebo, the mean reported effect on spasticity was not significantly different between control and treatment groups[15]. However, patient-reported spasticity was reduced, agreeing with earlier studies that showed subjective improvements in spasticity related to MS[14,15]. However, in a 12-mo follow up study of 657 patients, Δ9-THC was reported to have a significant effect on the objective measures of spasticity[16]. Several later studies have also shown variable results depending on cannabis preparation, dosing regime and patient numbers, throwing into doubt the use of clinical measures of spasticity.

At present, the large number of clinical trials in MS have not clarified whether cannabis is beneficial in MS but have thrown up questions about the rating of “spasticity”, route of delivery, source and dosing regime[10]. Despite these reservations, self-medicated use of cannabis continues. In a postal questionnaire involving the responses received from 110 MS patients in the South of England, 43% confirmed their use of cannabis with 68% of these patients specifically using cannabis to relieve the symptoms of MS[17]. Patients affirmed that their main reason for choosing to self-medicate with cannabis was due to pain and spasms, while a small proportion used cannabis for sleep related problems[17]. In a further study, over 90% of a cohort of 112 MS patients based in the United Kingdom and United States declared that self-medication with cannabis improved nocturnal pain, spasms and muscular pain[18].

A recent study showed that of the 572 MS patients enrolled in a clinical study, 272 (47.6%) responded to Sativex, an oral-mucosal spray that contains Δ9-THC and CBD, treatment within 4 wk with a response that was defined as > 20% decrease in spasticity[19]. A second phase of this study demonstrated that the cannabis extracts significantly reduced spasticity and the frequency of spasms while improving sleep quality over an extended 12-wk period, compared to placebo controls[19]. This has led to approval of the use of Sativex in the treatment of MS-related spasticity. These studies demonstrate the potential beneficial roles of cannabis use in MS patients, although in general its supply and use remains illegal.

ENDOCANNABINOID SYSTEM

The discovery of the endogenous cannabinoid (endocannabinoid) system that naturally exists in the body and responds to cannabinoids to exert their effects has aided research into the therapeutic utility of cannabinoids. This has fuelled the search for alternative modes of delivery into the human body, rather than the traditional method of “smoking”. The endocannabinoid system consists of at least two families of lipid signalling molecules (the N-acyl ethanolamines and the monoacyl-glycerols), multiple enzymes in the biosynthesis and degradation of these lipids, as well as two G-protein coupled receptors [cannabinoid receptor type-1 (CB1) and CB2] (reviewed by[10]). CB1 is mainly expressed in the CNS while CB2 is predominantly found in leukocytes, while an increasing number of potential ligands and endocannabinoid degradation molecules are being isolated[20]. Several studies have highlighted the involvement of this system in regulating neurotransmission and its ability to prevent excessive neurotransmitter release, consistent with a capacity to provide symptomatic relief in MS[21,22].

Endocannabinoids are produced on demand and are retrogradely transported across the postsynaptic membrane to engage with CB1 receptors, suppressing neurotransmitter release. The pharmacology of endocannabinoids is rapidly evolving with the discovery of new cannabinoid mimetics and novel CB receptor interactions that include orphan receptors and other CB receptors being proposed[23-26]. All these receptor functions are either sensitive to, or are regulated by CB receptors and culminate in neuropathic pain and inflammation, suggesting that novel drug targets based around these proteins might be useful in treating neuroinflammation and neuropathic pain in different neurological conditions. In addition, activation of the CB receptor inhibits adenylate cyclase that then reduces the levels of the second messenger cyclic adenosine monophosphate (cAMP), thus regulating cellular mechanisms such as cell fate[27]. CB receptor activation also inhibits voltage-dependent Ca2+ channels and activates inwardly rectifying K+ channels, a process that underlies CB-induced depression of excitatory neurotransmission[28,29]. Thus, the CB system is a potentially useful target for exploitation in neurodegenerative diseases such as MS.

AXONAL DAMAGE IN MS

Although MS is defined as an inflammatory demyelinating disease of the CNS, axonal loss is also a key feature of the disease. In MS patients, analysis of their spinal cord lesions suggested that the permanent loss in function was not primarily due to demyelination but due to axonal loss[30]. Axonal loss is generally associated with inflammatory macrophage infiltration but is variable in MS lesions and can be severe in certain cases. For example, axonal density is reduced by 60%-70% in actively demyelinating lesions and is characterised by the presence of axonal spheroids, endbulbs, or focal accumulation of proteins[31-33]. Although shadow plaques appear after injury that is consistent with an attempt to remyelinate axons, ongoing axonal injury is present[34]. This suggests that during the early stages of remyelination axons are more susceptible to damage, a process that is related to the patterns of Na+ channels at the widened surfaces of the nodes of Ranvier[35]. Axonal injury is also present in normal appearing white matter[36,37] and may occur as a result of secondary Wallerian degeneration[38]. However, this is not the only mechanism of axonal damage since two distinct patterns are observed: one that takes place within demyelinated lesions and correlates with lesional activity, while the other is diffuse axonal injury that is associated with inflammation and can additionally affect non-demyelinated nerve fibres[33].

Axonal injury in MS is also selective to the size of axon fibres. Small calibre axons are more prone to injury compared to thick axons and may relate to thin axons requiring a higher energy demand in terms of their critical mass of mitochondria[39]. Like MS, widespread axonal damage is also seen in experimental allergic encephalomyelitis (EAE)[34,40]. However, experimental models of MS reveal different mechanisms of axonal injury and there is currently no agreement for which mechanism is relevant to MS patients. Axonal injury mechanisms may involve T-lymphocytes and antibodies as part of the adaptive immune response as well as components of the innate immune system, driven by macrophages and microglia. For example, axonal injury can be driven by an antigen-specific cytotoxic T cell response, induced in neurons and glia by the expression of pro-inflammatory cytokines, leading to neuronal death and axonal transection[41-43].

Another mechanism of axonal damage relies on the production of auto-antibodies against cell surface molecules in neurons and axons. A subset of MS patients were described that mounted an antibody response against neurofascin, which is expressed on axons and oligodendrocyte processes at the nodes of Ranvier[44]. Systemic injection of neurofascin during EAE exacerbates the clinical symptoms of the disease with severe levels of axonal injury within lesions[44]. These observations suggest that auto-antibodies can directly mediate axonal damage. In MS lesions, axon damage is closely linked to the presence of macrophages and microglia that are in intimate contact with axons[3,5] and are known to produce a number of cytotoxic molecules including reactive oxygen and nitric oxide intermediates[33]. The expression of these molecules, especially inducible nitric oxide synthase from macrophages, correlates with areas of axonal injury in acute EAE[45]. In summary, axonal damage is a feature of MS and EAE and correlates with functional deficits. EAE models demonstrate that axonal damage occurs through a variety of mechanisms. However, recent reports have highlighted the role of mitochondrial injury and subsequent energy failure, induced by oxygen and nitric oxide free radicals as a possible mechanism of axonal injury. Understanding of the pathway to axon injury will aid in the discovery of new molecules for therapeutic intervention.

CANNABINOIDS IN MS

There is an abundance of evidence suggesting that MS patients gain symptomatic relief from cannabis extracts[46]. For example, Sativex, an oral-mucosal spray containing Δ9-THC and CBD is anti-spasmodic and analgesic in MS patients[47], while neuropathic pain associated with MS is relieved by dronabinol, an oral preparation of Δ9-THC analog[15]. Meta-analysis has also revealed that CB-based preparations are superior in the treatment of MS-related neuropathic pain than the placebo, confirming their beneficial effects in symptomatic relief[48]. This is consistent with the animal model of MS, EAE where treatment with exogenous cannabinoids controlled spasticity in chronic relapsing EAE models[49].

Modulation of the endocannabinoid system is also apparent in MS and EAE such that brain levels of CB receptors are downregulated in EAE while plasma levels of endocannabinoids are increased in MS patients[50,51]. Synthetic CB, HU-211, reduced the clinical severity of acute EAE in female Lewis rats as well as reducing inflammatory cell infiltration into the CNS[52], while the WIN55, 212-2, a CB1 and CB2 receptor agonist reduces T cell differentiation and hence reduces EAE severity in Theiler’s murine encephalomyelitis virus-induced demyelinating disease, a mouse model of chronic-progressive MS[53], suggesting a key involvement of the CB receptors in the pathogenesis of EAE. Induction of EAE in CB1 receptor-deficient mice causes rapid and progressively more neurodegeneration than in wild-type counterparts[54].

In our highly cited study on the role of cannabinoids in EAE, reported that the cannabinoid system was neuroprotective during EAE since CB1-deficient mice poorly tolerated inflammatory and excitotoxic insults and showed significant accumulation of neurodegeneration after EAE[55]. We induced chronic relapsing EAE (CREAE) in wild type ABH, CB1 gene (Cnr1)-deficient and congenic ABH. Cnr1+/+ mice with mouse spinal cord homogenate emulsified in complete Freund’s adjuvant on days 0 and 7 and monitored clinical disease progression over time. Mice developed characteristic paralytic disease episodes followed by remission with an increasing amount of residual deficit[49,56]. Whilst disease induction in both CB1-deficient and CB1 wild-type mice were similar, CB1-deficient mice exhibited significantly higher levels of residual deficit. This deficit was quantitated in an open-field activity chamber and confirmed that CB1-deficient mice displayed significantly more immobility and paresis than wild type mice, accumulating significantly more axonal damage after relapses. CB1-deificent mice also developed spasticity after only a single attack, which is not seen in wild type mice until after three to four relapses of disease[49].

Numerous mechanisms cause neuronal death and axonal damage in EAE, including the influx of toxic ions such as Ca2+ and caspase-3-mediated apoptosis[57]. CB1-deficient mice demonstrated significantly lower levels of active caspase-3 during acute EAE compared with wild type mice, while caspase-3 was detected in dying axons, consistent with that observed in MS[3]. These results suggested that the elevated neurodegeneration in CB1-deficient mice may be due to caspase-3-mediated apoptosis and axonal damage and hence agonism of the CB1 receptor pathway is neuroprotective and may control neurological symptoms such as tremor and spasticity[49].

We also investigated whether glutamate toxicity can be regulated by cannabinoids[55]. Glutamate excitotoxity causes neuronal damage in both MS and EAE. For example, the glutamate antagonist, amantadine, reduces the relapse rate in MS patients[58], while elevated glutamate levels have been observed in cerebrospinal fluid from MS patients[59]. In EAE, the enhanced levels of glutamate agonists may result in aberrant astrocyte function, since activated astrocytes normally regulate glutamate levels through enzymes such as glutamate dehydrogenase and glutamine synthetase, both of which are down-regulated during EAE[60,61]. The amount of CNS glutamate is also affected by abnormal changes in neuronal and glial glutamate transporters, all of which raise the levels of glutamate in the CNS during EAE and ultimately lead to the synthesis of mediators responsible for neuronal dysfunction[59,62-64]. We reported that after in vitro stimulation of N-methyl-D-aspartic (NMDA) receptors in cerebellar granule cells, there was significantly more neuronal Ca2+ influx in CB1-deficient mice than in wild type controls suggesting that the cannabinoid receptors may tonically regulate Ca2+ influx. The NMDA receptor antagonist MK-801 took longer to reduce Ca2+ back to basal levels in CB1-deficient mice than in congenic controls while CB1 agonism with CP55, 940 inhibited NMDA-induced Ca2+ influx in wild type mice but had no effect on CB1-deficient mice. These results suggest that Ca2+ is not only dysregulated in the absence of CB1 receptors but that postsynaptic control of NMDA-receptor activation is also compromised. These in vitro results were confirmed by in vitro injections of kainic acid, a specific agonist of the kainate receptor that mimics the effects of glutamate, since injection of kainic acid in CB1-deficient mice induced seizures and mortality within 10 min, while no effect of kainic acid was observed in wild type or congenic wild type control mice despite using 50-fold higher doses. Therefore, CB1-receptors clearly regulate ionotropic glutamate receptor activity, leading to enhanced susceptibility to excitotoxic damage in CB1-deficient mice.

Furthermore, we reported that CB1 agonists protected mice against the consequences of CNS inflammation in models of experimental allergic uveitis (EAU)[55]. EAU is an inflammatory disease of the eye and after sensitization with for example, interphotoreceptor retinal binding peptide in B10. RIII mice, the neuroretina is completely destroyed within 14-16 d[65]. CB1 receptor agonism with R(+)-WIN-55,212-2 and Δ9-THC both significantly inhibited photoreceptor damage without affecting inflammatory infiltrates suggesting that agonism of CB1 is neuroprotective. Taken together, the results of our study demonstrated that cannabinoids protect against the neurodegenerative events in models of MS and EAU.

CANNABINOID-1 RECEPTOR-MEDIATED NEUROPROTECTION IN MODELS OF MS

A feature of MS is neuronal loss, and in MS patients, loss of spinal cord axons correlate with neurological disability together with reduced N-acetyl aspartate levels in chronic MS patients[32]. Although axonal loss occurs early during the progression of MS[66], once a threshold of 15%-35% axonal loss in mice has been reached, permanent disability results[67,68]. In CB1-deficient mice, significant axonal loss is evident after a single acute attack of EAE, suggesting that the mere presence of CB1 is itself neuroprotective[55]. In EAE and MS, the presence of axonal damage correlates with inflammation that produces a range of neurotoxic agents such as glutamate, cytokines as well as creating oxidative stress in the environment of the CNS and damaging the blood-brain barrier[3,5,52,69-72].

Cannabinoids, however, can protect against acute hypoxia, excitotoxicity, oxidative and traumatic insults both in vitro and in vitro [73-76]. Cannabinoids can also inhibit both pre- and post-synaptic glutamate induced calcium responses and thus inhibit neurotoxicity[21,55,76], an effect that we showed to be CB1-dependent[55]. In accord with our observations, CB1-deficient mice were more susceptible to NMDA and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA)/kainite glutamate receptor excitotoxicity[55], however, Δ9-THC and CBD protected against NMDA-, AMPA- and kainite-agonist-induced cell death[77,78]. Delta-9-THC also protected retinal neurons from death induced by peroxynitrite-mediated NMDA-induced toxicity[79]. These observations demonstrate a clear neuroprotective role of cannabinoids in MS.

CANNABINOID-2-MEDIATED EFFECTS IN MODELS OF MS

Although much of the focus in MS is devoted to CB1 receptor pharmacology, the recognition that CB2 receptors possess immunomodulatory properties has led to an increased focus on CB2 receptors as potential therapeutic targets. Unlike CB1 receptors, activation of the CB2 receptor is not psychoactive and therefore targeting CB2 receptors with selective agonists is a promising therapeutic avenue that is immunomodulatory without being psychoactive. For example, early indications came from experiments that showed that the administration of WIN55212-2, which functions as a CB1 and CB2 receptor agonist, attenuated the progression of EAE in C57BL/6 mice immunized with myelin oligodendrocyte-derived glycoprotein35-55 (MOG35-55)[80]. Furthermore, a selective antagonist of the CB1 did not modulate the protective effect of WIN55212-2 but a selective antagonist of the CB2 receptor blocked the effects of WIN55212-2. This led to the suggestion that the protective effects of WIN55212-2 was mediated through the CB2 receptor[80]. However, later studies showed that the CB1 receptor might play a neuroprotective role in the latter stages of EAE[49,54].

Other highly selective CB2 receptor agonists such as O-1966 are also useful in the fight against MS since they do not produce psychoactive effects, determined by its low affinity to CB1 receptors[81]. Administration of O-1966 in a chronic (C57BL/6/MOG), relapsing-remitting and an adoptive transfer model attenuated disease progression and improved motor function[82]. In addition, encephalitogenic T cells derived from CB2-deficient mice were shown to be more aggressive in terms of CNS infiltration and increased severity of EAE, despite displaying similar levels of proliferation, apoptosis and cytokine production in the spleen as wild-type T cells[83]. Moreover, treatment of mice with CB2 receptor agonists attenuate white cell trafficking across the blood-brain barrier while dendritic cells differentiated in the presence of selective CB2 receptor agonist and inhibited T cell proliferation and shifted cytokine responses from inflammatory to anti-inflammatory molecules[82]. Recently, it has been shown that high concentrations of IFN-γ disrupts P2X7 purinergic receptor signalling and thus inhibit the neuroprotective effects of endogenous cannabinoids[84]. Therefore, inhibiting IFN-γ by exogenous CB2 agonists represents an obvious therapy to prevent the disruption of P2X7 signalling.

VALUE OF ANIMAL MODELS IN THE STUDY OF MULTIPLE SCLEROSIS

The validity of EAE as a model of MS is a topic of active debate with some researchers contenting that it is unsuitable due to its inability to mimic some of the pathological, immunologic and chronic features of MS. For example, EAE is usually monophasic whereas MS displays chronic relapsing features. Histological and magnetic resonance imaging data demonstrate axonal and cortical damage in MS but not in some models of EAE[85]. The extravasation of red blood cells into the CNS of swiss jim lambert mice and Lewis rats with EAE are not typical of MS[86,87], while encephalitogenic regions associated with myelin basic protein (MBP) or proteolipid protein normally activate more CD4+ than CD8+ T cells but in inflammatory MS, CD8+ T cell predominate[88-90]. However, there are many other immunological differences between mouse and human MS and these must be overcome if greater levels of success in drug development for human MS are to be achieved[91].

Several other points are worth considering in terms of the use of EAE as an animal model: (1) EAE provides little insight into the progression of MS in terms of the small amounts of demyelinated axons in EAE compared to MS while mice with EAE rarely exhibit ongoing functional deterioration that MS patients often display[92,93]; (2) The use of C57BL/6 mice in EAE studies limits the investigation of the mechanism of relapsing-remitting forms that more commonly affect MS patients (Ransohoff et al[93], 2002); (3) Treatment with factors that exert neurobiological effects also impact on immune and inflammatory cells and thus making results difficult to interpret (Ransohoff et al[93], 2002); and (4) EAE is generally generated using antigens that affect CD4+ T cells while CD8+ T cells that predominate in MS lesions are overlooked[92,94]. Likewise, the role of B cells is largely neglected despite recent data that demonstrate their importance in the pathogenesis of MS[95,96].

In summary, EAE has a long history in the fight against MS, however, its predictive value for treatment efficacy is poor. Nevertheless, it is widely used as a first-line animal model of MS and has provided mechanistic insights into the neuroinflammatory aspects of MS.

LIMITATIONS OF ANIMAL MODELS OF MS

One of the biggest limitations on the use of EAE as a model of MS is the fact that disease has to be induced with complete Freund’s adjuvant and heat-inactivated Mycobacterium tuberculosis rather than mimicking a spontaneous disease like MS. This leaves very little room for disease pathways and fails to represent the complexity of disease inducing mechanisms in MS. Demyelination, a feature of MS is also not obvious in all EAE models while the time course for disease manifestation may be days, EAE more closely resembles post-infectious acute demyelinating events[97]. In contrast, MS develops over years with patients presenting with more protracted epitope spreading than that observed in EAE mice[98]. There are also many other immunological differences between EAE and MS that need consideration in the development of potential therapies for MS.

Therapeutic developments from EAE have translated poorly to human MS. For example, only a few molecules that showed efficacy in EAE have been successful in MS trials. One of these molecules is Glatiramer acetate, a synthetic amino acid copolymer originally designed to mimic encephalitogenic MBP, but instead suppresses EAE by other mechanisms and reduced MS relapses by 30%[99,100]. However, the efficacy of Glatiramer acetate has been questioned by a systematic Cochrane review which calls into question the use of Glatiramer acetate in MS[101]. Tysabri (Natalizumab) and Gilenya (fingolimod) are the only two other drugs that have been licence for use in human MS. Natalizumab binds to the α4 subunit of α4β1 and α4β7 integrins and blocks binding to their endothelial receptors (VCAP-1 and mucosal addressin-cell adhesion molecule 1, thereby attenuating inflammation and ongoing inflammation[102,103]. Fingolimod is a sphingosine-1-phosphate-receptor modulator that prevents egress of lymphocytes from lymph nodes and significantly improved relapse rates compared to placebo controls[104]. At present therefore, it remains unclear why pre-clinical EAE studies predict treatment efficacy in human MS so poorly, however, EAE is still an important first-line model system in the development of new treatments for MS.

BETTER ANIMAL MODELS OF MULTIPLE SCLEROSIS

To make greater progress, better animals models of MS need to be considered when testing new drugs. One fact that to be borne in mind is the fact that MS is a heterogeneous disease in terms of genetics, environmental effects, disease course, pathological treatments and treatment responsiveness[105]. Currently the majority of experiments are performed in inbred strains while clinically, the molecular mechanisms that determine the efficacy during prolonged follow-up of hundreds of patients are far more complex. Thus, it is likely that genetic differences account for some of the inter-patient differences in clinical efficacy of various drugs. Improved animal models may take into account the therapeutic effect of drugs in more than one animal model, treatments to be instigated after disease onset, long-term disease periods, use spontaneous disease models and incorporate human risk. The use of partially humanized mouse models to address the genetic and disease variability are a step in the right direction towards developing better therapeutics for MS[106].

SUMMARY AND FUTURE PROSPECTS

Studies have shown that antagonism of the CB1 receptor pathway contributes to neuronal damage in CREAE and the relative worsening of tremor and spasticity. The addition of exogenous CB1 agonists derived from cannabis afforded significant neuroprotection from the consequences of inflammatory CNS disease in EAE and EAU models. Although clear neuroprotective benefits of cannabinoids have been demonstrated, the unwanted psychotropic effects need to be addressed. The adverse psychotropic effects, its role in appetite, pain and cognition together with the observation that the CB1 receptor is downregulated in some neurodegenerative diseases limits the usefulness of cannabinoids in the treatment of MS. Alternative treatments may be more useful and includes the development of drugs based on CBD, the non-psychoactive part of cannabis that possess anti-inflammatory and anti-oxidant properties. Furthermore, the CB2 receptor is being recognised as a potential target since it regulates neuroinflammation and neurogenesis while being non-psychoactive.

In summary, the endocannabinoid system exerts multiple actions and may be useful in the development of therapies to treat neurodegenerative diseases. However, the biggest challenge remains, namely the development of drugs that lack the adverse psychoactive side effects.

Footnotes

P- Reviewer: Mellick GD S- Editor: Song XX L- Editor: A E- Editor: Liu SQ

References
1.  Compston A, Coles A. Multiple sclerosis. Lancet. 2002;359:1221-1231.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1514]  [Cited by in F6Publishing: 1433]  [Article Influence: 65.1]  [Reference Citation Analysis (0)]
2.  Compston A, Coles A. Multiple sclerosis. Lancet. 2008;372:1502-1517.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3360]  [Cited by in F6Publishing: 3339]  [Article Influence: 208.7]  [Reference Citation Analysis (0)]
3.  Trapp BD, Peterson J, Ransohoff RM, Rudick R, Mörk S, Bö L. Axonal transection in the lesions of multiple sclerosis. N Engl J Med. 1998;338:278-285.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2859]  [Cited by in F6Publishing: 2673]  [Article Influence: 102.8]  [Reference Citation Analysis (0)]
4.  Coles AJ, Wing MG, Molyneux P, Paolillo A, Davie CM, Hale G, Miller D, Waldmann H, Compston A. Monoclonal antibody treatment exposes three mechanisms underlying the clinical course of multiple sclerosis. Ann Neurol. 1999;46:296-304.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Ferguson B, Matyszak MK, Esiri MM, Perry VH. Axonal damage in acute multiple sclerosis lesions. Brain. 1997;120:393-399.  [PubMed]  [DOI]  [Cited in This Article: ]
6.  Bjartmar C, Wujek JR, Trapp BD. Axonal loss in the pathology of MS: consequences for understanding the progressive phase of the disease. J Neurol Sci. 2003;206:165-171.  [PubMed]  [DOI]  [Cited in This Article: ]
7.  Smith KJ, Kapoor R, Hall SM, Davies M. Electrically active axons degenerate when exposed to nitric oxide. Ann Neurol. 2001;49:470-476.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Chaturvedi GN, Tiwari SK, Rai NP. Medicinal use of opium and cannabis in medieval India. Indian J Hist Sci. 1981;16:31-35.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Pertwee RG. Cannabinoids and multiple sclerosis. Pharmacol Ther. 2002;95:165-174.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Rog DJ. Cannabis-based medicines in multiple sclerosis--a review of clinical studies. Immunobiology. 2010;215:658-672.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
11.  Gaoni Y, Mechoulam R. The isolation and structure of delta-1-tetrahydrocannabinol and other neutral cannabinoids from hashish. J Am Chem Soc. 1971;93:217-224.  [PubMed]  [DOI]  [Cited in This Article: ]
12.  Gowran A, Noonan J, Campbell VA. The multiplicity of action of cannabinoids: implications for treating neurodegeneration. CNS Neurosci Ther. 2011;17:637-644.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 80]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
13.  Smith PF. Cannabinoids for the treatment of multiple sclerosis: no smoke without fire. Expert Rev Neurother. 2003;3:327-334.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
14.  Killestein J, Hoogervorst EL, Reif M, Blauw B, Smits M, Uitdehaag BM, Nagelkerken L, Polman CH. Immunomodulatory effects of orally administered cannabinoids in multiple sclerosis. J Neuroimmunol. 2003;137:140-143.  [PubMed]  [DOI]  [Cited in This Article: ]
15.  Zajicek J, Fox P, Sanders H, Wright D, Vickery J, Nunn A, Thompson A. Cannabinoids for treatment of spasticity and other symptoms related to multiple sclerosis (CAMS study): multicentre randomised placebo-controlled trial. Lancet. 2003;362:1517-1526.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 546]  [Cited by in F6Publishing: 475]  [Article Influence: 22.6]  [Reference Citation Analysis (0)]
16.  Zajicek JP, Sanders HP, Wright DE, Vickery PJ, Ingram WM, Reilly SM, Nunn AJ, Teare LJ, Fox PJ, Thompson AJ. Cannabinoids in multiple sclerosis (CAMS) study: safety and efficacy data for 12 months follow up. J Neurol Neurosurg Psychiatry. 2005;76:1664-1669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 195]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
17.  Chong MS, Wolff K, Wise K, Tanton C, Winstock A, Silber E. Cannabis use in patients with multiple sclerosis. Mult Scler. 2006;12:646-651.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Consroe P, Musty R, Rein J, Tillery W, Pertwee R. The perceived effects of smoked cannabis on patients with multiple sclerosis. Eur Neurol. 1997;38:44-48.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Novotna A, Mares J, Ratcliffe S, Novakova I, Vachova M, Zapletalova O, Gasperini C, Pozzilli C, Cefaro L, Comi G. A randomized, double-blind, placebo-controlled, parallel-group, enriched-design study of nabiximols* (Sativex(®) ), as add-on therapy, in subjects with refractory spasticity caused by multiple sclerosis. Eur J Neurol. 2011;18:1122-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 298]  [Cited by in F6Publishing: 283]  [Article Influence: 21.8]  [Reference Citation Analysis (0)]
20.  Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham M, Mackie K, Martin BR. International Union of Pharmacology. XXVII. Classification of cannabinoid receptors. Pharmacol Rev. 2002;54:161-202.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Wilson RI, Nicoll RA. Endogenous cannabinoids mediate retrograde signalling at hippocampal synapses. Nature. 2001;410:588-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1070]  [Cited by in F6Publishing: 1153]  [Article Influence: 50.1]  [Reference Citation Analysis (0)]
22.  Kreitzer AC, Carter AG, Regehr WG. Inhibition of interneuron firing extends the spread of endocannabinoid signaling in the cerebellum. Neuron. 2002;34:787-796.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Thakur GA, Tichkule R, Bajaj S, Makriyannis A. Latest advances in cannabinoid receptor agonists. Expert Opin Ther Pat. 2009;19:1647-1673.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 69]  [Cited by in F6Publishing: 74]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
24.  Brown AJ. Novel cannabinoid receptors. Br J Pharmacol. 2007;152:567-575.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 268]  [Cited by in F6Publishing: 290]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
25.  Sharir H, Abood ME. Pharmacological characterization of GPR55, a putative cannabinoid receptor. Pharmacol Ther. 2010;126:301-313.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 157]  [Article Influence: 11.2]  [Reference Citation Analysis (0)]
26.  Nevalainen T, Irving AJ. GPR55, a lysophosphatidylinositol receptor with cannabinoid sensitivity. Curr Top Med Chem. 2010;10:799-813.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Miyamoto N, Tanaka R, Zhang N, Shimura H, Onodera M, Mochizuki H, Hattori N, Urabe T. Crucial role for Ser133-phosphorylated form of cyclic AMP-responsive element binding protein signaling in the differentiation and survival of neural progenitors under chronic cerebral hypoperfusion. Neuroscience. 2009;162:525-536.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 25]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
28.  Hampson AJ, Grimaldi M, Lolic M, Wink D, Rosenthal R, Axelrod J. Neuroprotective antioxidants from marijuana. Ann N Y Acad Sci. 2000;899:274-282.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Nicholson RA, Liao C, Zheng J, David LS, Coyne L, Errington AC, Singh G, Lees G. Sodium channel inhibition by anandamide and synthetic cannabimimetics in brain. Brain Res. 2003;978:194-204.  [PubMed]  [DOI]  [Cited in This Article: ]
30.  Kornek B, Lassmann H. Axonal pathology in multiple sclerosis. A historical note. Brain Pathol. 1999;9:651-656.  [PubMed]  [DOI]  [Cited in This Article: ]
31.  Mews I, Bergmann M, Bunkowski S, Gullotta F, Brück W. Oligodendrocyte and axon pathology in clinically silent multiple sclerosis lesions. Mult Scler. 1998;4:55-62.  [PubMed]  [DOI]  [Cited in This Article: ]
32.  Bjartmar C, Kidd G, Mörk S, Rudick R, Trapp BD. Neurological disability correlates with spinal cord axonal loss and reduced N-acetyl aspartate in chronic multiple sclerosis patients. Ann Neurol. 2000;48:893-901.  [PubMed]  [DOI]  [Cited in This Article: ]
33.  Lassmann H. Axonal and neuronal pathology in multiple sclerosis: what have we learnt from animal models. Exp Neurol. 2010;225:2-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 121]  [Cited by in F6Publishing: 126]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
34.  Kornek B, Storch MK, Weissert R, Wallstroem E, Stefferl A, Olsson T, Linington C, Schmidbauer M, Lassmann H. Multiple sclerosis and chronic autoimmune encephalomyelitis: a comparative quantitative study of axonal injury in active, inactive, and remyelinated lesions. Am J Pathol. 2000;157:267-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 695]  [Cited by in F6Publishing: 651]  [Article Influence: 27.1]  [Reference Citation Analysis (0)]
35.  Smith KJ. Axonal protection in multiple sclerosis--a particular need during remyelination. Brain. 2006;129:3147-3149.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 35]  [Cited by in F6Publishing: 36]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
36.  Kutzelnigg A, Lucchinetti CF, Stadelmann C, Brück W, Rauschka H, Bergmann M, Schmidbauer M, Parisi JE, Lassmann H. Cortical demyelination and diffuse white matter injury in multiple sclerosis. Brain. 2005;128:2705-2712.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1241]  [Cited by in F6Publishing: 1204]  [Article Influence: 63.4]  [Reference Citation Analysis (0)]
37.  Frischer JM, Bramow S, Dal-Bianco A, Lucchinetti CF, Rauschka H, Schmidbauer M, Laursen H, Sorensen PS, Lassmann H. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132:1175-1189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 935]  [Cited by in F6Publishing: 985]  [Article Influence: 65.7]  [Reference Citation Analysis (0)]
38.  Evangelou N, Konz D, Esiri MM, Smith S, Palace J, Matthews PM. Regional axonal loss in the corpus callosum correlates with cerebral white matter lesion volume and distribution in multiple sclerosis. Brain. 2000;123:1845-1849.  [PubMed]  [DOI]  [Cited in This Article: ]
39.  Evangelou N, Konz D, Esiri MM, Smith S, Palace J, Matthews PM. Size-selective neuronal changes in the anterior optic pathways suggest a differential susceptibility to injury in multiple sclerosis. Brain. 2001;124:1813-1820.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Linington C, Bradl M, Lassmann H, Brunner C, Vass K. Augmentation of demyelination in rat acute allergic encephalomyelitis by circulating mouse monoclonal antibodies directed against a myelin/oligodendrocyte glycoprotein. Am J Pathol. 1988;130:443-454.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Neumann H, Cavalié A, Jenne DE, Wekerle H. Induction of MHC class I genes in neurons. Science. 1995;269:549-552.  [PubMed]  [DOI]  [Cited in This Article: ]
42.  Medana IM, Gallimore A, Oxenius A, Martinic MM, Wekerle H, Neumann H. MHC class I-restricted killing of neurons by virus-specific CD8+ T lymphocytes is effected through the Fas/FasL, but not the perforin pathway. Eur J Immunol. 2000;30:3623-3633.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
43.  Medana I, Martinic MA, Wekerle H, Neumann H. Transection of major histocompatibility complex class I-induced neurites by cytotoxic T lymphocytes. Am J Pathol. 2001;159:809-815.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 191]  [Cited by in F6Publishing: 199]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
44.  Mathey EK, Derfuss T, Storch MK, Williams KR, Hales K, Woolley DR, Al-Hayani A, Davies SN, Rasband MN, Olsson T. Neurofascin as a novel target for autoantibody-mediated axonal injury. J Exp Med. 2007;204:2363-2372.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 293]  [Cited by in F6Publishing: 311]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
45.  Aboul-Enein F, Weiser P, Höftberger R, Lassmann H, Bradl M. Transient axonal injury in the absence of demyelination: a correlate of clinical disease in acute experimental autoimmune encephalomyelitis. Acta Neuropathol. 2006;111:539-547.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 57]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
46.  Lakhan SE, Rowland M. Whole plant cannabis extracts in the treatment of spasticity in multiple sclerosis: a systematic review. BMC Neurol. 2009;9:59.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
47.  Rog DJ, Nurmikko TJ, Young CA. Oromucosal delta9-tetrahydrocannabinol/cannabidiol for neuropathic pain associated with multiple sclerosis: an uncontrolled, open-label, 2-year extension trial. Clin Ther. 2007;29:2068-2079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 137]  [Cited by in F6Publishing: 146]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
48.  Iskedjian M, Bereza B, Gordon A, Piwko C, Einarson TR. Meta-analysis of cannabis based treatments for neuropathic and multiple sclerosis-related pain. Curr Med Res Opin. 2007;23:17-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 124]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
49.  Baker AM, Grekova MC, Richert JR. EAE susceptibility in FVB mice. J Neurosci Res. 2000;61:140-145.  [PubMed]  [DOI]  [Cited in This Article: ]
50.  Jean-Gilles L, Feng S, Tench CR, Chapman V, Kendall DA, Barrett DA, Constantinescu CS. Plasma endocannabinoid levels in multiple sclerosis. J Neurol Sci. 2009;287:212-215.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 76]  [Cited by in F6Publishing: 80]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
51.  Centonze D, Bari M, Rossi S, Prosperetti C, Furlan R, Fezza F, De Chiara V, Battistini L, Bernardi G, Bernardini S. The endocannabinoid system is dysregulated in multiple sclerosis and in experimental autoimmune encephalomyelitis. Brain. 2007;130:2543-2553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 133]  [Cited by in F6Publishing: 150]  [Article Influence: 8.8]  [Reference Citation Analysis (0)]
52.  Achiron A, Miron S, Lavie V, Margalit R, Biegon A. Dexanabinol (HU-211) effect on experimental autoimmune encephalomyelitis: implications for the treatment of acute relapses of multiple sclerosis. J Neuroimmunol. 2000;102:26-31.  [PubMed]  [DOI]  [Cited in This Article: ]
53.  Croxford JL. Therapeutic potential of cannabinoids in CNS disease. CNS Drugs. 2003;17:179-202.  [PubMed]  [DOI]  [Cited in This Article: ]
54.  Maresz K, Pryce G, Ponomarev ED, Marsicano G, Croxford JL, Shriver LP, Ledent C, Cheng X, Carrier EJ, Mann MK. Direct suppression of CNS autoimmune inflammation via the cannabinoid receptor CB1 on neurons and CB2 on autoreactive T cells. Nat Med. 2007;13:492-497.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 263]  [Cited by in F6Publishing: 278]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
55.  Pryce G, Ahmed Z, Hankey DJ, Jackson SJ, Croxford JL, Pocock JM, Ledent C, Petzold A, Thompson AJ, Giovannoni G. Cannabinoids inhibit neurodegeneration in models of multiple sclerosis. Brain. 2003;126:2191-2202.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 245]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
56.  Baker D, O’Neill JK, Gschmeissner SE, Wilcox CE, Butter C, Turk JL. Induction of chronic relapsing experimental allergic encephalomyelitis in Biozzi mice. J Neuroimmunol. 1990;28:261-270.  [PubMed]  [DOI]  [Cited in This Article: ]
57.  Ahmed Z, Doward AI, Pryce G, Taylor DL, Pocock JM, Leonard JP, Baker D, Cuzner ML. A role for caspase-1 and -3 in the pathology of experimental allergic encephalomyelitis : inflammation versus degeneration. Am J Pathol. 2002;161:1577-1586.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 47]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
58.  Plaut GS. Effectiveness of amantadine in reducing relapses in multiple sclerosis. J R Soc Med. 1987;80:91-93.  [PubMed]  [DOI]  [Cited in This Article: ]
59.  Stover JF, Pleines UE, Morganti-Kossmann MC, Kossmann T, Lowitzsch K, Kempski OS. Neurotransmitters in cerebrospinal fluid reflect pathological activity. Eur J Clin Invest. 1997;27:1038-1043.  [PubMed]  [DOI]  [Cited in This Article: ]
60.  Hardin-Pouzet H, Krakowski M, Bourbonnière L, Didier-Bazes M, Tran E, Owens T. Glutamate metabolism is down-regulated in astrocytes during experimental allergic encephalomyelitis. Glia. 1997;20:79-85.  [PubMed]  [DOI]  [Cited in This Article: ]
61.  Rothstein JD. Neurobiology. Bundling up excitement. Nature. 2000;407:141, 143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 9]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
62.  Ohgoh M, Hanada T, Smith T, Hashimoto T, Ueno M, Yamanishi Y, Watanabe M, Nishizawa Y. Altered expression of glutamate transporters in experimental autoimmune encephalomyelitis. J Neuroimmunol. 2002;125:170-178.  [PubMed]  [DOI]  [Cited in This Article: ]
63.  Piani D, Frei K, Do KQ, Cuénod M, Fontana A. Murine brain macrophages induced NMDA receptor mediated neurotoxicity in vitro by secreting glutamate. Neurosci Lett. 1991;133:159-162.  [PubMed]  [DOI]  [Cited in This Article: ]
64.  Smith QR. Transport of glutamate and other amino acids at the blood-brain barrier. J Nutr. 2000;130:1016S-1022S.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Hankey DJ, Nickerson JM, Donoso LA, Lightman SL, Baker D. Experimental autoimmune uveoretinitis in mice (Biozzi ABH and NOD) expressing the autoimmune-associated H-2A(g7) molecule: identification of a uveitogenic epitope. J Neuroimmunol. 2001;118:212-222.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Filippi M, Rocca MA. MRI aspects of the “inflammatory phase” of multiple sclerosis. Neurol Sci. 2003;24 Suppl 5:S275-S278.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
67.  Confavreux C, Vukusic S, Moreau T, Adeleine P. Relapses and progression of disability in multiple sclerosis. N Engl J Med. 2000;343:1430-1438.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 895]  [Cited by in F6Publishing: 823]  [Article Influence: 34.3]  [Reference Citation Analysis (0)]
68.  Wujek JR, Bjartmar C, Richer E, Ransohoff RM, Yu M, Tuohy VK, Trapp BD. Axon loss in the spinal cord determines permanent neurological disability in an animal model of multiple sclerosis. J Neuropathol Exp Neurol. 2002;61:23-32.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Bolton C, Paul C. MK-801 limits neurovascular dysfunction during experimental allergic encephalomyelitis. J Pharmacol Exp Ther. 1997;282:397-402.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Pitt D, Werner P, Raine CS. Glutamate excitotoxicity in a model of multiple sclerosis. Nat Med. 2000;6:67-70.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 648]  [Cited by in F6Publishing: 670]  [Article Influence: 27.9]  [Reference Citation Analysis (0)]
71.  Koprowski H, Zheng YM, Heber-Katz E, Fraser N, Rorke L, Fu ZF, Hanlon C, Dietzschold B. In vivo expression of inducible nitric oxide synthase in experimentally induced neurologic diseases. Proc Natl Acad Sci USA. 1993;90:3024-3027.  [PubMed]  [DOI]  [Cited in This Article: ]
72.  Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, Garren H, Langer-Gould A, Strober S, Cannella B, Allard J. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med. 2002;8:500-508.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1293]  [Cited by in F6Publishing: 1270]  [Article Influence: 57.7]  [Reference Citation Analysis (0)]
73.  Shen M, Thayer SA. Cannabinoid receptor agonists protect cultured rat hippocampal neurons from excitotoxicity. Mol Pharmacol. 1998;54:459-462.  [PubMed]  [DOI]  [Cited in This Article: ]
74.  Nagayama T, Sinor AD, Simon RP, Chen J, Graham SH, Jin K, Greenberg DA. Cannabinoids and neuroprotection in global and focal cerebral ischemia and in neuronal cultures. J Neurosci. 1999;19:2987-2995.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Sinor AD, Irvin SM, Greenberg DA. Endocannabinoids protect cerebral cortical neurons from in vitro ischemia in rats. Neurosci Lett. 2000;278:157-160.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  Abood ME, Rizvi G, Sallapudi N, McAllister SD. Activation of the CB1 cannabinoid receptor protects cultured mouse spinal neurons against excitotoxicity. Neurosci Lett. 2001;309:197-201.  [PubMed]  [DOI]  [Cited in This Article: ]
77.  Hampson AJ, Grimaldi M, Axelrod J, Wink D. Cannabidiol and (-)Delta9-tetrahydrocannabinol are neuroprotective antioxidants. Proc Natl Acad Sci USA. 1998;95:8268-8273.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Marsicano G, Moosmann B, Hermann H, Lutz B, Behl C. Neuroprotective properties of cannabinoids against oxidative stress: role of the cannabinoid receptor CB1. J Neurochem. 2002;80:448-456.  [PubMed]  [DOI]  [Cited in This Article: ]
79.  El-Remessy AB, Khalil IE, Matragoon S, Abou-Mohamed G, Tsai NJ, Roon P, Caldwell RB, Caldwell RW, Green K, Liou GI. Neuroprotective effect of (-)Delta9-tetrahydrocannabinol and cannabidiol in N-methyl-D-aspartate-induced retinal neurotoxicity: involvement of peroxynitrite. Am J Pathol. 2003;163:1997-2008.  [PubMed]  [DOI]  [Cited in This Article: ]
80.  Ni X, Geller EB, Eppihimer MJ, Eisenstein TK, Adler MW, Tuma RF. Win 55212-2, a cannabinoid receptor agonist, attenuates leukocyte/endothelial interactions in an experimental autoimmune encephalomyelitis model. Mult Scler. 2004;10:158-164.  [PubMed]  [DOI]  [Cited in This Article: ]
81.  Wiley JL, Beletskaya ID, Ng EW, Dai Z, Crocker PJ, Mahadevan A, Razdan RK, Martin BR. Resorcinol derivatives: a novel template for the development of cannabinoid CB(1)/CB(2) and CB(2)-selective agonists. J Pharmacol Exp Ther. 2002;301:679-689.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Zhang M, Martin BR, Adler MW, Razdan RJ, Kong W, Ganea D, Tuma RF. Modulation of cannabinoid receptor activation as a neuroprotective strategy for EAE and stroke. J Neuroimmune Pharmacol. 2009;4:249-259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
83.  Dittel BN. Direct suppression of autoreactive lymphocytes in the central nervous system via the CB2 receptor. Br J Pharmacol. 2008;153:271-276.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
84.  Whiting P, Harbord R, Main C, Deeks JJ, Filippini G, Egger M, Sterne JA. Accuracy of magnetic resonance imaging for the diagnosis of multiple sclerosis: systematic review. BMJ. 2006;332:875-884.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 44]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
85.  Sriram S, Yao SY, Stratton C, Moses H, Narayana PA, Wolinsky JS. Pilot study to examine the effect of antibiotic therapy on MRI outcomes in RRMS. J Neurol Sci. 2005;234:87-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 15]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
86.  Raine CS, Traugott U, Nussenblatt RB, Stone SH. Optic neuritis and chronic relapsing experimental allergic encephalomyelitis: relationship to clinical course and comparison with multiple sclerosis. Lab Invest. 1980;42:327-335.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Forge JK, Pedchenko TV, LeVine SM. Iron deposits in the central nervous system of SJL mice with experimental allergic encephalomyelitis. Life Sci. 1998;63:2271-2284.  [PubMed]  [DOI]  [Cited in This Article: ]
88.  Booss J, Esiri MM, Tourtellotte WW, Mason DY. Immunohistological analysis of T lymphocyte subsets in the central nervous system in chronic progressive multiple sclerosis. J Neurol Sci. 1983;62:219-232.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Hauser SL, Bhan AK, Gilles F, Kemp M, Kerr C, Weiner HL. Immunohistochemical analysis of the cellular infiltrate in multiple sclerosis lesions. Ann Neurol. 1986;19:578-587.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 270]  [Cited by in F6Publishing: 266]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
90.  Lassmann H, Ransohoff RM. The CD4-Th1 model for multiple sclerosis: a critical [correction of crucial] re-appraisal. Trends Immunol. 2004;25:132-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 167]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
91.  Friese MA, Montalban X, Willcox N, Bell JI, Martin R, Fugger L. The value of animal models for drug development in multiple sclerosis. Brain. 2006;129:1940-1952.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 112]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
92.  Ransohoff RM. Animal models of multiple sclerosis: the good, the bad and the bottom line. Nat Neurosci. 2012;15:1074-1077.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 226]  [Cited by in F6Publishing: 252]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
93.  Ransohoff RM, Howe CL, Rodriguez M. Growth factor treatment of demyelinating disease: at last, a leap into the light. Trends Immunol. 2002;23:512-516.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Huseby ES, Liggitt D, Brabb T, Schnabel B, Ohlén C, Goverman J. A pathogenic role for myelin-specific CD8(+) T cells in a model for multiple sclerosis. J Exp Med. 2001;194:669-676.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Hauser SL, Waubant E, Arnold DL, Vollmer T, Antel J, Fox RJ, Bar-Or A, Panzara M, Sarkar N, Agarwal S. B-cell depletion with rituximab in relapsing-remitting multiple sclerosis. N Engl J Med. 2008;358:676-688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1742]  [Cited by in F6Publishing: 1695]  [Article Influence: 105.9]  [Reference Citation Analysis (0)]
96.  Berer K, Wekerle H, Krishnamoorthy G. B cells in spontaneous autoimmune diseases of the central nervous system. Mol Immunol. 2011;48:1332-1337.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 32]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
97.  Steinman L. Assessment of animal models for MS and demyelinating disease in the design of rational therapy. Neuron. 1999;24:511-514.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Vanderlugt CL, Miller SD. Epitope spreading in immune-mediated diseases: implications for immunotherapy. Nat Rev Immunol. 2002;2:85-95.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 603]  [Cited by in F6Publishing: 601]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
99.  Teitelbaum D, Meshorer A, Hirshfeld T, Arnon R, Sela M. Suppression of experimental allergic encephalomyelitis by a synthetic polypeptide. Eur J Immunol. 1971;1:242-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 334]  [Cited by in F6Publishing: 345]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
100.  Johnson KP, Brooks BR, Cohen JA, Ford CC, Goldstein J, Lisak RP, Myers LW, Panitch HS, Rose JW, Schiffer RB. Copolymer 1 reduces relapse rate and improves disability in relapsing-remitting multiple sclerosis: results of a phase III multicenter, double-blind placebo-controlled trial. The Copolymer 1 Multiple Sclerosis Study Group. Neurology. 1995;45:1268-1276.  [PubMed]  [DOI]  [Cited in This Article: ]
101.  Munari L, Lovati R, Boiko A. Therapy with glatiramer acetate for multiple sclerosis. Cochrane Database Syst Rev. 2004;CD004678.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 29]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
102.  Yednock TA, Cannon C, Fritz LC, Sanchez-Madrid F, Steinman L, Karin N. Prevention of experimental autoimmune encephalomyelitis by antibodies against alpha 4 beta 1 integrin. Nature. 1992;356:63-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1272]  [Cited by in F6Publishing: 1250]  [Article Influence: 39.1]  [Reference Citation Analysis (0)]
103.  Polman CH, O’Connor PW, Havrdova E, Hutchinson M, Kappos L, Miller DH, Phillips JT, Lublin FD, Giovannoni G, Wajgt A. A randomized, placebo-controlled trial of natalizumab for relapsing multiple sclerosis. N Engl J Med. 2006;354:899-910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2326]  [Cited by in F6Publishing: 2279]  [Article Influence: 126.6]  [Reference Citation Analysis (0)]
104.  Kappos L, Comi G, Panitch H, Oger J, Antel J, Conlon P, Steinman L. Induction of a non-encephalitogenic type 2 T helper-cell autoimmune response in multiple sclerosis after administration of an altered peptide ligand in a placebo-controlled, randomized phase II trial. The Altered Peptide Ligand in Relapsing MS Study Group. Nat Med. 2000;6:1176-1182.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 409]  [Cited by in F6Publishing: 386]  [Article Influence: 16.1]  [Reference Citation Analysis (0)]
105.  Lassmann H, Brück W, Lucchinetti C. Heterogeneity of multiple sclerosis pathogenesis: implications for diagnosis and therapy. Trends Mol Med. 2001;7:115-121.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Gregersen JW, Holmes S, Fugger L. Humanized animal models for autoimmune diseases. Tissue Antigens. 2004;63:383-394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 34]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]