Downloaded from https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3142952/

 

Mt Sinai J Med. Author manuscript; available in PMC 2012 Mar 1.

 

Published in final edited form as:

Mt Sinai J Med. 2011 Mar; 78(2): 231–243.

doi:  10.1002/msj.20246

PMCID: PMC3142952

NIHMSID: NIHMS306828

Axonal Damage in Multiple Sclerosis

Jeffery D. Haines, PhD,1 Matilde Inglese, MD, PhD,2 and Patrizia Casaccia, MD, PhD1

Author information ► Copyright and License information ►

 

The publisher's final edited version of this article is available at Mt Sinai J Med

See other articles in PMC that cite the published article.

 

Go to:

Abstract

Multiple sclerosis is a debilitating disease of the central nervous system that has been characteristically classified as an immune-mediated destruction of myelin, the protective coating on nerve fibers. Although the mechanisms responsible for the immune attack to central nervous system myelin have been the subject of intense investigation, more recent studies have focused on the neurodegenerative component, which is cause of clinical disability in young adults and appears to be only partially controlled by immunomodulatory therapies. Here, we review distinct, but not mutually exclusive, mechanisms of pathogenesis of axonal damage in multiple sclerosis patients that are either consequent to long-term demyelination or independent from it. We propose that the complexity of axonal degeneration and the heterogeneity of the underlying pathogenetic mechanisms should be taken into consideration for the design of targeted therapeutic intervention.

Keywords: axon, demyelination, multiple sclerosis, myelin, neurodegeneration, neuroimaging, oligodendrocyte, pathogenesis

Multiple sclerosis (MS) is a debilitating neurological disorder of young adults with a high prevalence in North America and Europe, afflicting almost 2.5 million individuals worldwide,1,2 and with an incidence rate that continues to rise.3 Approximately 85% of MS patients begin with a relapsing-remitting (RRMS) course of the disease, characterized by clinically debilitating events followed by return to baseline.4 After a variable period of time, the majority of RRMS patients develop a secondary progressive (SPMS) form of the disease, which is characterized by persistent and progressive disease advancement without remissions.5 In a small proportion of MS patients (10%–15%), the disease advances rapidly without periods of remittance and is termed primary progressive MS (PPMS).6 Historically, axonal damage has been recognized as a histopathological hallmark of MS since the very early descriptions of the disease by Charcot in the late 1800s.7 More recent clinical, histopathological, and neuroimaging evidence has shed new light on these early findings and supported the concept that the debilitating disease course and long-term disability in MS patients was consequent to axonal loss possibly consequent to demyelination.8,9 The features of axonal damage in MS, however, were similar to those detected in other neurological diseases lacking demyelination, such as amyotrophic lateral sclerosis (ALS), and suggested the possibility that axonal damage in MS might be concurrent to demyelination, but not necessarily consequent to myelin destruction.10 This review discusses the hypothesis that axonal damage underlying the debilitating disease progression observed in MS patients may be caused by mechanisms other than long-term demyelination.

Axonal damage in multiple sclerosis might be concurrent to demyelination, but not necessarily consequent to myelin destruction.

Go to:

 

HISTOPATHOLOGICAL EVIDENCE OF AXONAL DAMAGE IN MULTIPLE SCLEROSIS

Morphological and Histopathological Characteristics of Axonal Damage

Axonal damage is a common feature of many neurodegenerative diseases. The morphological characteristics of early axonal damage include the presence of varicosities and spheroid structures,11 which are associated with impaired transport of proteins and organelles along the axon.12 Disrupted axonal transport can be detected by immunohistochemistry using antibodies specific for the amyloid precursor protein (APP) as a marker. In physiological conditions APP is rapidly transported along the axons and cannot be easily detected by immunoreactivity.13,14 In case of axonal dysfunction and altered transport, in contrast, APP accumulates in localized axonal enlargements that become immunoreactive and that can be detected in active, remyelinating, and inactive MS lesions.15,16 An additional marker of axonal damage is the nonphosphorylated form of neurofilament heavy chain (NFH). In healthy axons, NFH is phosphorylated and this correlates with fast axonal transport.17 In compromised axons, in contrast, the nonphosphorylated form of NFH, which can be identified by SMI-32 immunoreactivity, can be used as maker of neurodegeneration,18 ALS,19 and MS.20

Evidence for Axonal Damage as Consequence of Primary Demyelination

A correlation between axonal damage and demyelination has been determined from histopathological examination of MS postmortem tissue. Histopathological analysis of early MS lesions showed that most axonal transections occur during the process of active demyelination.21,22 Furthermore, regional axonal loss in the corpus callosum correlated with the cerebral white matter (WM) lesion volume distribution and was suggested to be a result of degenerated axons transected in demyelinated lesions.23 Axons in MS lesions also stained with antibodies specific for nonphosphorylated NFH and total axonal loss were correlated to the degree of inflammatory demyelination,21 suggesting that neurodegeneration occurs as the consequence of myelin loss.

Further evidence supporting this notion was the observation that the extent of remyelination extended lifespan of mice and provided a protective effect on axons.24 Perhaps the most striking evidence to substantiate axonal degeneration as a consequence of demyelination was obtained from animal models of MS, including experimental autoimmune encephalomyelitis (EAE) and cuprizone-induced demyelination. In EAE, animals are immunized with antigenic myelin extracts or peptides (eg, myelin-oligodendrocyte glycoprotein [MOG]), which elicit an immune T cell–mediated disease characterized by demyelination.25,26 After long-term demyelination in EAE, axonal loss is observed in rats immunized with MOG22 and in guinea pigs,27 substantiating the concept that axonal damage occurs following the loss of myelin support. Axonal loss can also be detected in the cuprizone-induced model of toxic demyelination in aged mice,28 which have less-efficient remyelination compared with young mice.29

Evidence in Favor of Hypothesis That Axonal Damage Can Occur Independently of Demyelination

The hypothesis that axonal damage in MS may occur independently of chronic demyelination has been suggested by several studies that will be reviewed in this manuscript. This section discusses recent neuropathological findings on gray matter (GM) lesions, meningeal infiltrates detected in human brain samples, and experimental evidence collected in genetically manipulated mouse mutants. The first evidence supporting the existence of alternative mechanisms of pathogenesis for axonal damage in MS were the description of GM lesions, characterized by neuronal loss and dendritic atrophy30 and the presence of axonal abnormalities in areas devoid of ongoing demyelination and thereby designated as normal appearing white matter (NAWM). Gray matter lesions differed from WM lesions3133 and were characterized by a different composition of the inflammatory cell infiltrate34,35 and less-prominent antibody complement activation compared with WM tracts.36 Gray matter lesions have been correlated with cortical thinning and were considered a predictive index of disability in MS patients,3739 including those with PPMS.40,41 In some studies, cortical demyelination in MS brain was detected during the late stages of disease pathology and interpreted as the consequence rather than the cause of neuronal loss.30 Neuronal cell death has also been detected in both cortical and thalamic lesion areas,34,42 and it could be mimicked by injection of neurofilament light chain (NFL) into mice, which produces a GM pathology with axonal loss and empty myelin sheaths, thereby suggesting an antibody-mediated primary axonal damage with a secondary involvement of myelin.43 This concept was supported by the detection of meningeal B-cell follicles in the brain of patients with a diagnosis of SPMS (but not PPMS) and by the correlation between the presence of these infiltrates and severe cortical pathology.44

The hypothesis that axonal damage in multiple sclerosis may occur independently of chronic demyelination has been suggested by several studies involving neuropathological findings on gray matter lesions and meningeal infiltrates in both human brain samples and genetically manipulated mouse mutants.

Very recent studies have further suggested a significant correlation between decreased axonal density in NAWM and diffuse parenchymal infiltration of major histocompatibility complex class II–positive and meningeal inflammatory infiltrates composed of CD3+ T cells in the cervical spinal cord of PPMS patients’ microglia.45 These intriguing data were consistent with other reports of inflammatory meningeal infiltrates in the brain of PPMS and SPMS patients with a high degree of axonal loss46 and provided a potential explanation for the axonal injury detected in the progressive course of the disease even in the absence of diffuse inflammatory infiltrates.47

A third line of evidence was provided by the description of axonal degeneration in mice with detectable myelin, even after genetic deletion of specific myelin proteins or oligodendrocyte components. Mice lacking the gene encoding for proteolipid protein (Plp), for instance, were characterized by progressive axonal degeneration.48 A similar phenotype was described for mice with genetic deletion of 2′, 3′-cyclic nucleotide phosphodiesterase,49 or of myelin-associated glycoprotein,50 which were characterized by relatively normal myelination and severe progressive axonal degeneration occurring with aging. Thus, even in the absence of morphological evidence of demyelination, microscopic imbalances of the oligodendrocyte-neuron unit may result in deregulation of energy metabolism, followed by destabilization and damage of the axon.51

Go to:

 

CLINICAL DATA SUPPORTING NEURONAL DAMAGE IN MS

Clinically, MS disease progression is measured by the Expanded Disability Status Scale (EDSS).52 Progressive exacerbation of the clinical symptoms is seen over time in SPMS patients and correlates most significantly with axonal damage.4 In addition, PPMS disease progression correlates with axonal loss and exhibits less-prominent inflammation than in SPMS.53,54 One of the most striking clinical observations is the fact that immunomodulatory therapies have been able to control the number of relapses,55 but patients continue to worsen clinically56 and show signs of cortical atrophy.57

Axonal damage in MS patients, as defined by spectroscopic measurements of metabolites, has also been correlated also with the progressive worsening of numerous symptoms including fatigue,58 cognitive dysfunction,59 and memory impairment.60 Very recently, cognitive impairment in MS patients was reported to be associated with GM atrophy,61 especially in the CA1 and CA3 region of the hippocampus.62 Together these data contribute to support the concept that the pathogenesis of neurodegeneration in MS may be independent of the immune system and blood-brain barrier permeability and involve other mechanisms and players.

Whereas immunomodulatory therapies have been able to control the number of relapses, patients continue to worsen clinically and show signs of cortical atrophy. The pathogenesis of neurodegeneration in multiple sclerosis may thus be independent of the immune system and blood-brain barrier permeability and involve other mechanisms and other players.

Go to:

BIOCHEMICAL DATA SUPPORTING NEURONAL DAMAGE IN MULTIPLE SCLEROSIS

Further supporting the notion that axonal injury occurs in MS, increased levels of axonal cytoskeletal proteins are found in the cerebral spinal fluid (CSF) of MS patients, including tubulin, actin, NFL,63 and tau.64 Interestingly, the detection of NFL in the CSF of MS patients positively correlates with EDSS score65 and its presence is not limited to the late stages of disease progression, but it is observed throughout the disease course, thereby suggesting that axonal damage may not only occur as the consequence of long-term demyelination.66 One intriguing hypothesis is that a primary neurodegenerative event generates axonal debris and possibly myelin degradation products that may then elicit the production of reactive autoantibodies that appear in the CSF, including those to myelin-specific proteins.67 However, we cannot exclude the possibility that the detection of autoantibodies against neuronal components could be the consequence of axonal damage and transections and thereby could be considered as marker of axonal integrity and disease progression.

Go to:

IMAGING DATA SUPPORTING NEURONAL DAMAGE IN MULTIPLE SCLEROSIS

Magnetic Resonance Imaging Studies

Magnetic resonance imaging (MRI) is widely used to diagnose MS and monitor disease progression by evaluating focal abnormalities in the CNS. Conventional MRI techniques have utility for the noninvasive measurement of WM lesions, including global and regional brain volume determinations.68 Advanced, quantitative MRI methods have the ability to detect early changes in the NAWM of MS patients, which provides clues to early disease pathogenesis.69 Longitudinal MRI studies in RRMS patients show that brain atrophy correlates positively with subsequent disability status.70,71 Thus, several studies correlate cortical thinning with MS disease severity but do not define whether thinning is just the consequence of axonal damage following long-term demyelination or occurring independently from it.57 New studies, reporting cortical and subcortical GM volume loss since the earliest stages of the disease, suggest that axonal damage may occur independently of demyelination.72 In a recent longitudinal study, Fisniku et al.38 evaluated tissue-specific atrophy in a cohort of 73 MS patients initially presenting with a clinically isolated syndrome (CIS) who were followed for 20 years, with clinical and MRI evaluations. The investigators noted that the extent of atrophy in the GM of MS patients was greater than that in WM areas, and that GM atrophy proved to be a stronger predictor of disability than focal WM lesion load and WM atrophy.38

New studies, reporting cortical and subcortical gray matter volume loss since the earliest stages of the disease, suggest that axonal damage may occur independently of demyelination.

With the advancement of powerful MRI techniques, the early occurrence of GM lesions is being further appreciated,73 although MRI techniques currently underestimate their number.74,75 Regional volume measurements by MRI have revealed smaller hippocampal CA1 regions in MS brains, and a subset of MS patients with depression had a smaller CA2-3/dentate gyrus volume.62

Another quantitative technique, diffusion MRI, measures the microscopic Brownian motion of water molecules. This motion is hindered by cellular structures such as cell membranes and the axonal cytoskeleton.76 Using diffusion MRI and applying field gradients in multiple directions, it is possible to infer the orientation of axons and reconstruct the pathways of the major WM bundles.77 Abnormalities in diffusivity patterns have been detected in focal MS lesions, NAWM, and GM. These abnormalities have been shown to correlate with physical disability78 and cognitive impairment in MS.79 Magnetic resonance imaging measurement of sodium ions in the plaques and NAWM of MS patients supports the hypothesis that demyelination results in eventual axonal damage. Sodium ion concentrations are higher in both acute and chronic lesions and in NAWM.80 This supports the idea that, following a demyelination event, sodium channel expression is increased and they redistribute along the axon in order to maintain nerve conduction,81 suggesting axons remain intact following demyelination.

Spectroscopic Measurement of Metabolites

The neuronal metabolite N-acetyl aspartate (NAA) is a measure of mitochondrial activity, which can be determined by proton magnetic resonance spectroscopy (1H-MRS).82,83 In the adult brain, NAA is present only in neurons and axons,84 so its measurement in vivo by 1H-MRS is useful to determine the extent of axonal damage/loss.85 In MS, NAA is decreased in both lesional areas and NAWM, which suggests that either mitochondrial dysfunction and/or axonal damage occurs also in regions devoid of active demyelination.82,86,87 Importantly, due to the high pathological specificity of NAA, its levels yield a better correlation with the degree of disability occurring in the presence or absence of demyelinating activity.8890 Correlative MRI-histological studies have shown that reduced NAA levels correlate with reduced axonal numbers in lesions of SPMS patients,91 and NAA concentrations are decreased in PPMS.92 Decreased NAA levels reverse as inflammation subsides following anti-inflammatory treatment with glatiramer acetate.93 Furthermore, a combination of NAA measurement, lesion imaging, and genetic analysis of MS patients with the DRB*1501 haplotype has been proposed as a method to stratify patients according to disease severity.94 Increased lactate levels in the CSF of MS patients have also been reported,95,96 whereas others have found decreased lactate levels in early stages of MS97 or throughout the MS disease course,98 suggesting its levels may fluctuate with disease progression or be indicative of disease heterogeneity.

The neuronal metabolite N-acetyl aspartate is a measure of mitochondrial activity, which can be determined by proton magnetic resonance spectroscopy. In multiple sclerosis, N-acetyl aspartate is decreased in both lesional areas and normal appearing white matter, which suggests that either mitochondrial dysfunction and/or axonal damage occurs also in regions devoid of active demyelination.

Importantly, due to the high pathological specificity of N-acetyl aspartate, its levels yield a better correlation with the degree of disability occurring in the presence or absence of demyelinating activity.

Other metabolite changes observed in NAWM of MS patients include the measurement of the excitatory neurotransmitter glutamate and of the glial-enriched metabolite myo-inositol.99 Notably, myo-inositol concentration was found to be elevated in the NAWM of patients with CIS suggestive of MS.100 Because myo-inositol is preferentially concentrated in glial cells,101 its increase may reflect astrocytosis and microglial activation. Because no correlation could be detected between NAWM myo-inositol levels and T2-lesion load, it is conceivable that the early detection of myo-inositol in CIS might reflect a relevant pathogenic process, which occurs independently from inflammatory demyelination.

The elevated levels of the excitatory neurotransmitter glutamate in acute MS lesions and NAWM but not in chronic MS lesions,102 together with the consistently decreased NAA levels in chronic lesions, suggest the possibility that excitotoxicity may trigger or be part of the mechanism leading to neurodegeneration in MS. Although these results will need to be confirmed in longitudinal studies assessing the predictive value of increased glutamate on the development of chronic lesions and brain atrophy, they are supported by studies in cultured neurons, where exposure to glutamate and inflammatory cytokines is sufficient to induce deficits in axonal transport and leads to the formation of localized varicosities and frank transections.103 Excessive glutamate release and impaired clearance may be cytotoxic to either neurons or oligodendrocytes.103105 The correlation of elevated glutamate levels and decreased NAA levels was also associated with the rs794185 noncoding single nucleotide polymorphism in a subset of MS patients with high brain volume loss and severe neurodegeneration.106 Thus, suggesting that glutamate may be part of the mechanisms that, within the context of an inflammatory environment might lead to axonal loss in MS.

The elevated levels of the excitatory neurotransmitter glutamate in acute multiple sclerosis lesions and normal appearing white matter but not in chronic multiple sclerosis lesions, together with the consistently decreased N-acetyl aspartate levels in chronic lesions, suggest the possibility that excitotoxicity may trigger or be part of the mechanism leading to neurodegeneration in multiple sclerosis.

Positron Emission Tomography

Positron emission tomography (PET) is a functional imaging technique which employs detection of radioligand tracers to clinically quantitate molecular processes of disease. In MS, PET imaging is used to correlate metabolic patterns to clinical symptoms, such as fatigue, cognitive impairment, and disability.107 Analysis of brain glucose metabolism in MS patients through PET shows increased glucose utilization, suggesting increased energy demand following demyelination.108 In MS, PET is also used to investigate microglial activation and inflammation as a marker of disease activity.109,110 Furthermore, PET imaging has shown decreased cortical cerebral metabolism in MS.111 Importantly, recent advancements have been made in radioligand development for PET imaging of demyelination/remyelination levels in animal models of MS.112 Therefore, PET imaging of neurodegeneration in relation to demyelination and remyelination holds promise for determining early pathological changes during the disease course of MS.

Go to:

 

POTENTIAL MECHANISMS OF DAMAGE

Axonal Damage Consequent to Demyelination

One of the potential mechanisms accountable for axonal loss following demyelination is Wallerian degeneration, whereby axons degenerate distal to the site of damage. Wallerian degeneration has been recognized as a contributor to axonal damage in MS.113

Reduced mitochondrial activity, due to increased energetic demand of demyelinated axons, can partially explain the decreased NAA levels observed in MS brain. Following demyelination, sodium channels, which are normally located at the nodes of Ranvier, become dispersed along the length of the demyelinated axon in order to restore nerve conduction.81,114 The increased number of sodium channels along the demyelinated axon requires a large quantity of sodium ions to be pumped back into the extracellular space by the energy-dependent Na+/K+-ATPase. Because adenosine triphosphate (ATP) concentrations are limited, accumulating sodium ions in the axoplasm results in impairment of Na+/K+-ATPase pump function. As a consequence, increased intra-cellular Na+ concentrations reverse the Na+/Ca2+ exchanger and permits Ca2+ entry into the cell. Further exacerbating this disrupted Ca2+ homeostasis in axons are reduced levels of the plasma membrane Ca2+ ATPase (PMCA2) efflux pump, which is seen in EAE,115,116 resulting in increased intracellular Ca2+ causing death of spinal cord neurons in vitro.117 In turn, increased axoplasmic concentration of Ca2+ activates proteases, impairs mitochondrial operation, depolymerizes microtubules, and compromises axonal transport.118,119

However, because axonal damage is detected also in areas devoid of demyelinating lesions and is barely controlled by current immunomodulatory therapies that limit the number and extent of demyelinating lesions, alternative pathogenetic mechanisms might come into play.

Axonal Damage Consequent to Direct Cytotoxic Attack

The exact mechanisms that underlie the progression of axonal damage are largely unknown. In vitro experimental evidence has shown CD4+ T cells can contribute to neurodegeneration in a mouse model of Parkinson’s Disease (PD).120 In addition, polyclonally activated CD4+/CD8+ effector cells align along axons and the soma of human neurons, causing neuronal cell death in vitro, but not death of oligodendrocytes or astrocytes.121 It has been proposed that loss of spinal motor neurons is the consequence of CD4+ and CD8+ T-cell infiltration in animal models of demyelination.122 However, the number of CD8+ T cells correlates with the extent of axonal damage in MS and the detection of CD8+ T cells in the brain,123 blood,124 and CSF125 of patients, in excess of CD4+ T cells,123 have suggested CD8+ T cells as important effectors of axonal damage. Indeed, accumulation of APP in damaged axons correlates with the number of macrophages and CD8+ T cells within MS lesions,15 CD8+ T cells closely interact with demyelinated axons in MS lesions126 and are capable of inducing direct damage to cultured murine neurons.127 Dying spinal motor neurons are surrounded by CD4+ and CD8+ T cells in EAE and postmortem MS tissue.122 Consistent with the proposed cytotoxic role of CD8+ T cells, depletion of CD4+ T cells in MS patients shows no improvement on relapse rate or inflammatory activity when visualized by MRI.128,129 Depletion of both CD8+ and CD4+ T cells, however, decreases relapse rate and new lesion formation but provides only little improvement of neurological symptoms.56,130 This suggests that CD8+ T cells are potential mediators of axonal damage. One possibility is that CD8+ T cells may cause damage indirectly, by targeting oligodendrocytes and myelin. For example, CD8+ T cells have been found to mediate the lysis of cultured murine oligodendrocytes131 and myelin in cerebellar slice cultures with axonal damage occurring as a bystander effect.132 The alternative possibility is that CD8+ T cells and possibly natural killer cells directly damage the axon due to the release of the cytolytic molecule perforin, a membrane pore-forming protein found in intracellular granules within these cells.133 The evidence to support the role for perforin in MS disease progression is substantiated from both the analysis of MS blood samples and the phenotype of mice with genetic ablation of the gene encoding for perforin. For example, high numbers of myelin basic protein (MBP)-reactive perforin mRNA expressing blood mononuclear cells (MNCs) are detected in MS patients134 and increased perforin expression is found in CD4+ T cells during MS disease exacerbation135 and correlates with the extent of brain lesions detected by MRI.136 Importantly, perforin-deficient mice are protected from axonal damage in the spinal cord and suggest that this might be a prominent mechanism of axonal degeneration.137

Axonal injury is therefore, at least in part, independent of demyelinating activity, and this concept may bear important implications for future therapeutic strategies aimed at preventing axonal loss.

Antibody/Complement-Mediated Lesion

We have previously discussed experimental evidence linking GM pathology, characterized by axonal loss and empty myelin sheaths to the injection of NFL into mice, thereby suggesting an antibody-mediated mechanism of damage.43 Furthermore, adoptive transfer of transiently expressed axonal glycoprotein 1 (TAG-1)-specific T cells into rats elicited a disease course with a preferential GM pathology, with WM demyelination only occurring after MOG antibody injection,138 thereby reflecting GM disease detected by early MRI measurements in MS patients. In further support of the hypothesis that neurodegeneration may underlie the pathogenesis of MS are also the findings of autoantibodies to neuronal and axonal antigens in the serum and CSF in a subset of MS patients, as well as the presence of meningeal B-cell infiltrates.44 The autoantibodies and antigens include neurofilament,139 axolemma enriched fractions,140 neurofascin,141 and contactin 2/TAG-1.138 However, some of these axonal or neuronal targets, namely intracytoplasmic proteins, may represent markers of injury rather than direct mediators of attack. Thus, neurodegeneration may underlie disease progression and WM demyelination, at least in a subset of patients.

Damage Caused by Dysfunctional Glial-Neuronal Crosstalk

The axonal degeneration detected in mice with deletion of genes encoding for myelin proteins suggested the existence of a bidirectional trophic support between myelin and neurons, affecting the long-term survival of neurons.51 For example, neurons and oligodendrocytes can exchange small metabolites necessary for myelin membrane lipid synthesis.142 The axonal degeneration detected in Plp-null mice, for instance, has been proposed to result from impaired transport of the nicotinamide adenine dinucleotide+-dependent deacetylase sirtuin 2, a cytoplasmic deacetylase implicated in axo-glial support. Similarly, ablation of a crucial peroxisomal enzyme in oligodendrocytes has been associated with dramatic axonal degeneration143 and the subsequent presence of inflammatory infiltrates.144 Thus, the loss of trophic support and deregulation of energy metabolism may destabilize metabolically isolated axons, resulting in their damage.51

Damage Caused by Exposure to Glutamate and Cytokines

We have discussed evidence linking inflammation to axonal damage even in the absence of demyelination. Meningeal infiltrates and diffuse microglial activation within the parenchyma have been associated with the release of cytokines and increased levels of the excitatory neurotransmitter glutamate. It is conceivable that these important mediators might directly modulate axonal function, within GM cortical areas (where the neurons are exposed to the meningeal infiltrates) or within normal appearing WM regions (characterized by diffuse microglial infiltration in the absence of demyelinating lesions). Recent evidence suggests that exposure of cultured neurons to glutamate and cytokines results in impaired axonal transport followed by axonal transections.103 This process was found to be mediated by a Ca2+-dependent export of the enzyme histone deacetylase 1 (HDAC1) from the nucleus to the axoplasm, with consequent disruption of mitochondrial and cargo transport along axons.103 Pharmacological inhibitors of HDAC1 (ie, MS-275), but not inhibitors of other cytosolic HDACs (ie, tubacin), were able to prevent the damage and partially restore axonal transport in neurons exposed to cytokines and glutamate, thereby suggesting protein acetylation as an important therapeutic target for axonal damage in demyelinating disorders.145

Exposure of cultured neurons to glutamate and cytokines results in impaired axonal transport followed by axonal transections. Pharmacological inhibitors of HDAC1 (ie, MS-275), but not inhibitors of other cytosolic HDACs (ie, tubacin), were able to prevent the damage and partially restore axonal transport in neurons exposed to cytokines and glutamate, thereby suggesting protein acetylation as an important therapeutic target for axonal damage in demyelinating disorders.

Go to:

 

CONCLUSION

Axonal damage is a prominent feature of MS responsible for long-term disability and functional deficits in those afflicted with the disease. In this review we outlined biochemical, clinical, neuropathological, and neuroimaging evidence in support of neurodegeneration as a direct consequence of demyelination or as an independently occurring concurrent event. Future studies should continue to address the molecular changes that occur to both axons and myelin in order to determine the specific sequence of events underlying the disease pathogenesis of MS. Novel therapies addressing the neurodegenerative component of the disease may hold clues for halting its progression and promoting functional recovery in those afflicted with MS.

Fig 1

Oligodendrocytes myelinate numerous axonal segments producing the compact myelin sheath that supports the long-term survival of neurons and their axonal segments (A). Axonal damage underlying MS disease progression may occur as a consequence of long-term ...

Go to:

Acknowledgments

This work is funded in part by a grant from the National Multiple Sclerosis Foundation (NMSS RG-4134/A9) and American Recovery and Reinvestment Act Funds (R01-NS42925-07S1 and NS42925-08) to PC.

Go to:

Footnotes

DISCLOSURES

Potential conflict of interest: Nothing to report.

Go to:

 

References

 

  1. Weinshenker BG. Epidemiology of multiple sclerosis. Neurol Clin. 1996;14:291–308. [PubMed]
  2. Noseworthy JH, Lucchinetti C, Rodriguez M, et al. Multiple sclerosis. N Engl J Med. 2000;343:938–952. [PubMed]
  3. Ebers GC. Environmental factors and multiple sclerosis. Lancet Neurol. 2008;7:268–277. [PubMed]
  4. Trapp BD, Nave KA. Multiple sclerosis: an immune or neurodegenerative disorder? Annu Rev Neurosci. 2008;31:247–269. [PubMed]
  5. Rovaris M, Confavreux C, Furlan R, et al. Secondary progressive multiple sclerosis: current knowledge and future challenges. Lancet Neurol. 2006;5:343–354. [PubMed]
  6. Thompson AJ, Polman CH, Miller DH, et al. Primary progressive multiple sclerosis. Brain. 1997;120(part 6):1085–1096. [PubMed]
  7. Kornek B, Lassmann H. Axonal pathology in multiple sclerosis: a historical note. Brain Pathol. 1999;9:651–656. [PubMed]
  8. Brück W. Inflammatory demyelination is not central to the pathogenesis of multiple sclerosis. J Neurol. 2005;252(suppl 5):v10–v15. [PubMed]
  9. Brück W. The pathology of multiple sclerosis is the result of focal inflammatory demyelination with axonal damage. J Neurol. 2005;252(suppl 5):v3–v9. [PubMed]
  10. De Vos KJ, Grierson AJ, Ackerley S, et al. Role of axonal transport in neurodegenerative diseases. Annu Rev Neurosci. 2008;31:151–173. [PubMed]
  11. Saxena S, Caroni P. Mechanisms of axon degeneration: from development to disease. Prog Neurobiol. 2007;83:174–191. [PubMed]
  12. Williamson TL, Cleveland DW. Slowing of axonal transport is a very early event in the toxicity of ALS-linked SOD1 mutants to motor neurons. Nat Neurosci. 1999;2:50–56. [PubMed]
  13. Koo EH, Sisodia SS, Archer DR, et al. Precursor of amyloid protein in Alzheimer disease undergoes fast anterograde axonal transport. Proc Natl Acad Sci U S A. 1990;87:1561–1565. [PMC free article] [PubMed]
  14. Terwel D, Dewachter I, Van Leuven F. Axonal transport, tau protein, and neurodegeneration in Alzheimer’s disease. Neuromolecular Med. 2002;2:151–165. [PubMed]
  15. Bitsch A, Schuchardt J, Bunkowski S, et al. Acute axonal injury in multiple sclerosis: correlation with demyelination and inflammation. Brain. 2000;123(part 6):1174–1183. [PubMed]
  16. Ferguson B, Matyszak MK, Esiri MM, et al. Axonal damage in acute multiple sclerosis lesions. Brain. 1997;120(part 3):393–399. [PubMed]
  17. Watson DF, Fittro KP, Hoffman PN, et al. Phosphorylation-related immunoreactivity and the rate of transport of neurofilaments in chronic 2,5-hexanedione intoxication. Brain Res. 1991;539:103–109. [PubMed]
  18. Su JH, Cummings BJ, Cotman CW. Plaque biogenesis in brain aging and Alzheimer’s disease. I. Progressive changes in phosphorylation states of paired helical filaments and neurofilaments. Brain Res. 1996;739:79–87. [PubMed]
  19. Nihei K, McKee AC, Kowall NW. Patterns of neuronal degeneration in the motor cortex of amyotrophic lateral sclerosis patients. Acta Neuropathol. 1993;86:55–64. [PubMed]
  20. Wegner C, Esiri MM, Chance SA, et al. Neocortical neuronal, synaptic, and glial loss in multiple sclerosis. Neurology. 2006;67:960–967. [PubMed]
  21. Trapp BD, Peterson J, Ransohoff RM, et al. Axonal transection in the lesions of multiple sclerosis. N Engl J Med. 1998;338:278–285. [PubMed]
  22. Kornek B, Storch MK, Weissert R, et al. 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. [PMC free article] [PubMed]
  23. Evangelou N, Konz D, Esiri MM, et al. Regional axonal loss in the corpus callosum correlates with cerebral white matter lesion volume and distribution in multiple sclerosis. Brain. 2000;123(part 9):1845–1849. [PubMed]
  24. Patrikios P, Stadelmann C, Kutzelnigg A, et al. Remyelination is extensive in a subset of multiple sclerosis patients [published correction appears in Brain 2007;130(part 3):879] Brain. 2006;129:3165–3172. [PubMed]
  25. Bolton C, Paul C. Glutamate receptors in neuro-inflammatory demyelinating disease. Mediators Inflamm. 2006;2006:93684. [PMC free article] [PubMed]
  26. Lassmann H. Recent neuropathological findings in MS–implications for diagnosis and therapy. J Neurol. 2004;251(suppl 4):IV2–IV5. [PubMed]
  27. Raine CS, Cross AH. Axonal dystrophy as a consequence of long-term demyelination. Lab Invest. 1989;60:714–725. [PubMed]
  28. Irvine KA, Blakemore WF. Age increases axon loss associated with primary demyelination in cuprizone-induced demyelination in C57BL/6 mice. J Neuroimmunol. 2006;175:69–76. [PubMed]
  29. Irvine KA, Blakemore WF. Remyelination protects axons from demyelination-associated axon degeneration. Brain. 2008;131(part 6):1464–1477. [PubMed]
  30. Siffrin V, Vogt J, Radbruch H, et al. Multiple sclerosis: candidate mechanisms underlying CNS atrophy. Trends Neurosci. 2010;33:202–210. [PubMed]
  31. Davies GR, Ramió-Torrentá L, Hadjiprocopis A, et al. Evidence for grey matter MTR abnormality in minimally disabled patients with early relapsing-remitting multiple sclerosis. J Neurol Neurosurg Psychiatry. 2004;75:998–1002. [PMC free article] [PubMed]
  32. Ramió-Torrentá L, Sastre-Garriga J, Ingle GT, et al. Abnormalities in normal appearing tissues in early primary progressive multiple sclerosis and their relation to disability: a tissue specific magnetisation transfer study. J Neurol Neurosurg Psychiatry. 2006;77:40–45. [PMC free article] [PubMed]
  33. De Stefano N, Matthews PM, Filippi M, et al. Evidence of early cortical atrophy in MS: relevance to white matter changes and disability. Neurology. 2003;60:1157–1162. [PubMed]
  34. Peterson JW, Bø L, Mørk S, et al. Transected neurites, apoptotic neurons, and reduced inflammation in cortical multiple sclerosis lesions. Ann Neurol. 2001;50:389–400. [PubMed]
  35. Bø L, Vedeler CA, Nyland H, et al. Intracortical multiple sclerosis lesions are not associated with increased lymphocyte infiltration. Mult Scler. 2003;9:323–331. [PubMed]
  36. Brink BP, Veerhuis R, Breij EC, et al. The pathology of multiple sclerosis is location-dependent: no significant complement activation is detected in purely cortical lesions. J Neuropathol Exp Neurol. 2005;64:147–155. [PubMed]
  37. Agosta F, Rovaris M, Pagani E, et al. Magnetization transfer MRI metrics predict the accumulation of disability 8 years later in patients with multiple sclerosis. Brain. 2006;129:2620–2627. [PubMed]
  38. Fisniku LK, Chard DT, Jackson JS, et al. Gray matter atrophy is related to long-term disability in multiple sclerosis [published correction appears in Ann Neurol 2009;65:232] Ann Neurol. 2008;64:247–254. [PubMed]
  39. Fisher E, Lee JC, Nakamura K, et al. Gray matter atrophy in multiple sclerosis: a longitudinal study. Ann Neurol. 2008;64:255–265. [PubMed]
  40. Rovaris M, Judica E, Gallo A, et al. Grey matter damage predicts the evolution of primary progressive multiple sclerosis at 5 years. Brain. 2006;129:2628–2634. [PubMed]
  41. Manfredonia F, Ciccarelli O, Khaleeli Z, et al. Normal-appearing brain t1 relaxation time predicts disability in early primary progressive multiple sclerosis. Arch Neurol. 2007;64:411–415. [PubMed]
  42. Cifelli A, Arridge M, Jezzard P, et al. Thalamic neuro-degeneration in multiple sclerosis. Ann Neurol. 2002;52:650–653. [PubMed]
  43. Huizinga R, Gerritsen W, Heijmans N, et al. Axonal loss and gray matter pathology as a direct result of autoimmunity to neurofilaments. Neurobiol Dis. 2008;32:461–470. [PubMed]
  44. Magliozzi R, Howell O, Vora A, et al. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain. 2007;130:1089–1104. [PubMed]
  45. Androdias G, Reynolds R, Chanal M, et al. Meningeal T cells associate with diffuse axonal loss in multiple sclerosis spinal cords. Ann Neurol. 2010;68:465–476. [PubMed]
  46. Frischer JM, Bramow S, Dal-Bianco A, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132:1175–1189. [PMC free article] [PubMed]
  47. Tallantyre EC, Bø L, Al-Rawashdeh O, et al. Greater loss of axons in primary progressive multiple sclerosis plaques compared to secondary progressive disease. Brain. 2009;132:1190–1199. [PubMed]
  48. Griffiths I, Klugmann M, Anderson T, et al. Axonal swellings and degeneration in mice lacking the major proteolipid of myelin. Science. 1998;280:1610–1613. [PubMed]
  49. Lappe-Siefke C, Goebbels S, Gravel M, et al. Disruption of Cnp1 uncouples oligodendroglial functions in axonal support and myelination. Nat Genet. 2003;33:366–374. [PubMed]
  50. Yin X, Crawford TO, Griffin JW, et al. Myelin-associated glycoprotein is a myelin signal that modulates the caliber of myelinated axons. J Neurosci. 1998;18:1953–1962. [PubMed]
  51. Nave KA. Myelination and the trophic support of long axons. Nat Rev Neurosci. 2010;11:275–283. [PubMed]
  52. Barkhof F. MRI in multiple sclerosis: correlation with expanded disability status scale (EDSS) Mult Scler. 1999;5:283–286. [PubMed]
  53. Revesz T, Kidd D, Thompson AJ, et al. A comparison of the pathology of primary and secondary progressive multiple sclerosis. Brain. 1994;117(part 4):759–765. [PubMed]
  54. Thompson AJ, Kermode AG, Wicks D, et al. Major differences in the dynamics of primary and secondary progressive multiple sclerosis. Ann Neurol. 1991;29:53–62. [PubMed]
  55. Confavreux C, Vukusic S, Moreau T, et al. Relapses and progression of disability in multiple sclerosis. N Engl J Med. 2000;343:1430–1438. [PubMed]
  56. Coles AJ, Wing MG, Molyneux P, et al. Monoclonal antibody treatment exposes three mechanisms underlying the clinical course of multiple sclerosis. Ann Neurol. 1999;46:296–304. [PubMed]
  57. Sailer M, Fischl B, Salat D, et al. Focal thinning of the cerebral cortex in multiple sclerosis. Brain. 2003;126:1734–1744. [PubMed]
  58. Tartaglia MC, Narayanan S, Francis SJ, et al. The relationship between diffuse axonal damage and fatigue in multiple sclerosis. Arch Neurol. 2004;61:201–207. [PubMed]
  59. Gadea M, Martínez-Bisbal MC, Marti-Bonmatí L, et al. Spectroscopic axonal damage of the right locus coeruleus relates to selective attention impairment in early stage relapsing-remitting multiple sclerosis. Brain. 2004;127:89–98. [PubMed]
  60. Pan JW, Krupp LB, Elkins LE, et al. Cognitive dysfunction lateralizes with NAA in multiple sclerosis. Appl Neuropsychol. 2001;8:155–160. [PubMed]
  61. Riccitelli G, Rocca MA, Pagani E, et al. Cognitive impairment in multiple sclerosis is associated to different patterns of gray matter atrophy according to clinical phenotype. Hum Brain Mapp. 2010 doi: 10.1002/hbm.21125. [PubMed] [Cross Ref]
  62. Gold SM, Kern KC, O’Connor MF, et al. Smaller cornu ammonis 2–3/dentate gyrus volumes and elevated cortisol in multiple sclerosis patients with depressive symptoms. Biol Psychiatry. 2010;68:553–559. [PMC free article] [PubMed]
  63. Semra YK, Seidi OA, Sharief MK. Heightened intrathecal release of axonal cytoskeletal proteins in multiple sclerosis is associated with progressive disease and clinical disability. J Neuroimmunol. 2002;122:132–139. [PubMed]
  64. Kapaki E, Paraskevas GP, Michalopoulou M, et al. Increased cerebrospinal fluid tau protein in multiple sclerosis. Eur Neurol. 2000;43:228–232. [PubMed]
  65. Lycke JN, Karlsson JE, Andersen O, et al. Neuro-filament protein in cerebrospinal fluid: a potential marker of activity in multiple sclerosis. J Neurol Neurosurg Psychiatry. 1998;64:402–404. [PMC free article] [PubMed]
  66. Malmeström C, Haghighi S, Rosengren L, et al. Neurofilament light protein and glial fibrillary acidic protein as biological markers in MS. Neurology. 2003;61:1720–1725. [PubMed]
  67. Tumani H, Hartung HP, Hemmer B, et al. BioMS Study Group. Cerebrospinal fluid biomarkers in multiple sclerosis. Neurobiol Dis. 2009;35:117–127. [PubMed]
  68. Tomassini V, Palace J. Multiple sclerosis lesions: insights from imaging techniques. Expert Rev Neurother. 2009;9:1341–1359. [PubMed]
  69. Vrenken H, Geurts JJ. Gray and normal-appearing white matter in multiple sclerosis: an MRI perspective. Expert Rev Neurother. 2007;7:271–279. [PubMed]
  70. Fisher E, Rudick RA, Simon JH, et al. Eight-year follow-up study of brain atrophy in patients with MS. Neurology. 2002;59:1412–1420. [PubMed]
  71. Rudick RA, Fisher E, Lee JC, et al. Use of the brain parenchymal fraction to measure whole brain atrophy in relapsing-remitting MS. Multiple Sclerosis Collaborative Research Group. Neurology. 1999;53:1698–1704. [PubMed]
  72. Ceccarelli A, Rocca MA, Pagani E, et al. A voxel-based morphometry study of grey matter loss in MS patients with different clinical phenotypes. Neuroimage. 2008;42:315–322. [PubMed]
  73. Geurts JJ, Pouwels PJ, Uitdehaag BM, et al. Intracortical lesions in multiple sclerosis: improved detection with 3D double inversion-recovery MR imaging. Radiology. 2005;236:254–260. [PubMed]
  74. Kidd D, Barkhof F, McConnell R, et al. Cortical lesions in multiple sclerosis. Brain. 1999;122(part 1):17–26. [PubMed]
  75. Geurts JJ, Bø L, Pouwels PJ, et al. Cortical lesions in multiple sclerosis: combined postmortem MR imaging and histopathology. AJNR Am J Neuroradiol. 2005;26:572–577. [PubMed]
  76. Le Bihan D, Mangin JF, Poupon C, et al. Diffusion tensor imaging: concepts and applications. J Magn Reson Imaging. 2001;13:534–546. [PubMed]
  77. Behrens TE, Johansen-Berg H, Woolrich MW, et al. Non-invasive mapping of connections between human thalamus and cortex using diffusion imaging. Nat Neurosci. 2003;6:750–757. [PubMed]
  78. Filippi M, Cercignani M, Inglese M, et al. Diffusion tensor magnetic resonance imaging in multiple sclerosis. Neurology. 2001;56:304–311. [PubMed]
  79. Rovaris M, Iannucci G, Falautano M, et al. Cognitive dysfunction in patients with mildly disabling relapsing-remitting multiple sclerosis: an exploratory study with diffusion tensor MR imaging. J Neurol Sci. 2002;195:103–109. [PubMed]
  80. Inglese M, Madelin G, Oesingmann N, et al. Brain tissue sodium concentration in multiple sclerosis: a sodium imaging study at 3 tesla. Brain. 2010;133:847–857. [PMC free article] [PubMed]
  81. Waxman SG. Axonal conduction and injury in multiple sclerosis: the role of sodium channels. Nat Rev Neurosci. 2006;7:932–941. [PubMed]
  82. Arnold DL, Matthews PM, Francis G, et al. Proton magnetic resonance spectroscopy of human brain in vivo in the evaluation of multiple sclerosis: assessment of the load of disease. Magn Reson Med. 1990;14:154–159. [PubMed]
  83. Wolinsky JS, Narayana PA, Fenstermacher MJ. Proton magnetic resonance spectroscopy in multiple sclerosis. Neurology. 1990;40:1764–1769. [PubMed]
  84. Simmons ML, Frondoza CG, Coyle JT. Immunocyto-chemical localization of N-acetyl-aspartate with monoclonal antibodies. Neuroscience. 1991;45:37–45. [PubMed]
  85. De Stefano N, Battaglini M, Smith SM. Measuring brain atrophy in multiple sclerosis. J Neuroimaging. 2007;17(suppl 1):10S–15S. [PubMed]
  86. De Stefano N, Narayanan S, Francis GS, et al. Evidence of axonal damage in the early stages of multiple sclerosis and its relevance to disability. Arch Neurol. 2001;58:65–70. [PubMed]
  87. Matthews PM, De Stefano N, Narayanan S, et al. Putting magnetic resonance spectroscopy studies in context: axonal damage and disability in multiple sclerosis. Semin Neurol. 1998;18:327–336. [PubMed]
  88. Grossman RI, Lenkinski RE, Ramer KN, et al. MR proton spectroscopy in multiple sclerosis. AJNR Am J Neuroradiol. 1992;13:1535–1543. [PubMed]
  89. Matthews PM, Francis G, Antel J, et al. Proton magnetic resonance spectroscopy for metabolic characterization of plaques in multiple sclerosis [published correction appears in Neurology 1991;41:1828] Neurology. 1991;41:1251–1256. [PubMed]
  90. Davie CA, Hawkins CP, Barker GJ, et al. Serial proton magnetic resonance spectroscopy in acute multiple sclerosis lesions. Brain. 1994;117(part 1):49–58. [PubMed]
  91. Bjartmar C, Kidd G, Mörk S, et al. 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]
  92. Suhy J, Rooney WD, Goodkin DE, et al. 1H MRSI comparison of white matter and lesions in primary progressive and relapsing-remitting MS. Mult Scler. 2000;6:148–155. [PMC free article] [PubMed]
  93. Khan O, Shen Y, Caon C, et al. Axonal metabolic recovery and potential neuroprotective effect of glatiramer acetate in relapsing-remitting multiple sclerosis. Mult Scler. 2005;11:646–651. [PubMed]
  94. Okuda DT, Srinivasan R, Oksenberg JR, et al. Genotype-Phenotype correlations in multiple sclerosis: HLA genes influence disease severity inferred by 1HMR spectroscopy and MRI measures. Brain. 2009;132:250–259. [PMC free article] [PubMed]
  95. Nicoli F, Vion-Dury J, Confort-Gouny S, et al. Cerebrospinal fluid metabolic profiles in multiple sclerosis and degenerative dementias obtained by high resolution proton magnetic resonance spectroscopy. C R Acad Sci III. 1996;319:623–631. [PubMed]
  96. Simone IL, Federico F, Trojano M, et al. High resolution proton MR spectroscopy of cerebrospinal fluid in MS patients. Comparison with biochemical changes in demyelinating plaques. J Neurol Sci. 1996;144:182–190. [PubMed]
  97. Fonalledas Perelló MA, Politi JV, Dallo Lizarraga MA, et al. The cerebrospinal fluid lactate is decreased in early stages of multiple sclerosis. P R Health Sci J. 2008;27:171–174. [PubMed]
  98. Aasly J, Gårseth M, Sonnewald U, et al. Cerebrospinal fluid lactate and glutamine are reduced in multiple sclerosis. Acta Neurol Scand. 1997;95:9–12. [PubMed]
  99. Gustafsson MC, Dahlqvist O, Jaworski J, et al. Low choline concentrations in normal-appearing white matter of patients with multiple sclerosis and normal MR imaging brain scans. AJNR Am J Neuroradiol. 2007;28:1306–1312. [PubMed]
  100. Fernando KT, McLean MA, Chard DT, et al. Elevated white matter myo-inositol in clinically isolated syndromes suggestive of multiple sclerosis. Brain. 2004;127:1361–1369. [PubMed]
  101. Brand A, Richter-Landsberg C, Leibfritz D. Multinuclear NMR studies on the energy metabolism of glial and neuronal cells. Dev Neurosci. 1993;15:289–298. [PubMed]
  102. Srinivasan R, Sailasuta N, Hurd R, et al. Evidence of elevated glutamate in multiple sclerosis using magnetic resonance spectroscopy at 3 T. Brain. 2005;128:1016–1025. [PubMed]
  103. Kim JY, Shen S, Dietz K, et al. HDAC1 nuclear export induced by pathological conditions is essential for the onset of axonal damage. Nat Neurosci. 2010;13:180–189. [PMC free article] [PubMed]
  104. Pitt D, Nagelmeier IE, Wilson HC, et al. Glutamate uptake by oligodendrocytes: Implications for excitotoxicity in multiple sclerosis. Neurology. 2003;61:1113–1120. [PubMed]
  105. Matute C. Oligodendrocyte NMDA receptors: a novel therapeutic target. Trends Mol Med. 2006;12:289–292. [PubMed]
  106. Baranzini SE, Srinivasan R, Khankhanian P, et al. Genetic variation influences glutamate concentrations in brains of patients with multiple sclerosis. Brain. 2010;133:2603–2611. [PMC free article] [PubMed]
  107. Kiferle L, Politis M, Muraro PA, et al. Positron emission tomography imaging in multiple sclerosis-current status and future applications. Eur J Neurol. 2011;18:226–231. [PubMed]
  108. Schiepers C, Van Hecke P, Vandenberghe R, et al. Positron emission tomography, magnetic resonance imaging and proton NMR spectroscopy of white matter in multiple sclerosis. Mult Scler. 1997;3:8–17. [PubMed]
  109. Banati RB, Newcombe J, Gunn RN, et al. The peripheral benzodiazepine binding site in the brain in multiple sclerosis: quantitative in vivo imaging of microglia as a measure of disease activity. Brain. 2000;123(part 11):2321–2337. [PubMed]
  110. Wilms H, Claasen J, Röhl C, et al. Involvement of benzodiazepine receptors in neuroinflammatory and neurodegenerative diseases: evidence from activated microglial cells in vitro. Neurobiol Dis. 2003;14:417–424. [PubMed]
  111. Blinkenberg M, Rune K, Jensen CV, et al. Cortical cerebral metabolism correlates with MRI lesion load and cognitive dysfunction in MS. Neurology. 2000;54:558–564. [PubMed]
  112. Wang Y, Wu C, Caprariello AV, et al. In vivo quantification of myelin changes in the vertebrate nervous system. J Neurosci. 2009;29:14663–14669. [PMC free article] [PubMed]
  113. Dziedzic T, Metz I, Dallenga T, et al. Wallerian degeneration: a major component of early axonal pathology in multiple sclerosis. Brain Pathol. 2010;20:976–985. [PubMed]
  114. Smith KJ. Sodium channels and multiple sclerosis: roles in symptom production, damage and therapy [published correction appears in Brain Pathol 2007;17:345] Brain Pathol. 2007;17:230–242. [PubMed]
  115. Nicot A, Ratnakar PV, Ron Y, et al. Regulation of gene expression in experimental autoimmune encephalomyelitis indicates early neuronal dysfunction. Brain. 2003;126:398–412. [PubMed]
  116. Nicot A, Kurnellas M, Elkabes S. Temporal pattern of plasma membrane calcium ATPase 2 expression in the spinal cord correlates with the course of clinical symptoms in two rodent models of autoimmune encephalomyelitis. Eur J Neurosci. 2005;21:2660–2670. [PMC free article] [PubMed]
  117. Kurnellas MP, Nicot A, Shull GE, et al. Plasma membrane calcium ATPase deficiency causes neuronal pathology in the spinal cord: a potential mechanism for neurodegeneration in multiple sclerosis and spinal cord injury. FASEB J. 2005;19:298–300. [PMC free article] [PubMed]
  118. Nave KA, Trapp BD. Axon-glial signaling and the glial support of axon function. Annu Rev Neurosci. 2008;31:535–561. [PubMed]
  119. Stys PK. White matter injury mechanisms. Curr Mol Med. 2004;4:113–130. [PubMed]
  120. Brochard V, Combadière B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J Clin Invest. 2009;119:182–192. [PMC free article] [PubMed]
  121. Giuliani F, Goodyer CG, Antel JP, et al. Vulnerability of human neurons to T cell-mediated cytotoxicity. J Immunol. 2003;171:368–379. [PubMed]
  122. Vogt J, Paul F, Aktas O, et al. Lower motor neuron loss in multiple sclerosis and experimental autoimmune encephalomyelitis. Ann Neurol. 2009;66:310–322. [PubMed]
  123. Babbe H, Roers A, Waisman A, et al. Clonal expansions of CD8(+) T cells dominate the T cell infiltrate in active multiple sclerosis lesions as shown by micromanipulation and single cell polymerase chain reaction. J Exp Med. 2000;192:393–404. [PMC free article] [PubMed]
  124. Crawford MP, Yan SX, Ortega SB, et al. High prevalence of autoreactive, neuroantigen-specific CD8+ T cells in multiple sclerosis revealed by novel flow cytometric assay. Blood. 2004;103:4222–4231. [PubMed]
  125. Skulina C, Schmidt S, Dornmair K, et al. Multiple sclerosis: brain-infiltrating CD8+ T cells persist as clonal expansions in the cerebrospinal fluid and blood. Proc Natl Acad Sci U S A. 2004;101:2428–2433. [PMC free article] [PubMed]
  126. Neumann H, Medana IM, Bauer J, et al. Cytotoxic T lymphocytes in autoimmune and degenerative CNS diseases. Trends Neurosci. 2002;25:313–319. [PubMed]
  127. Medana I, Martinic MA, Wekerle H, et al. Transection of major histocompatibility complex class I-induced neurites by cytotoxic T lymphocytes. Am J Pathol. 2001;159:809–815. [PMC free article] [PubMed]
  128. Van Oosten BW, Lai M, Hodgkinson S, et al. Treatment of multiple sclerosis with the monoclonal anti-CD4 antibody cM-T412: results of a randomized, double-blind, placebo-controlled, MR-monitored phase II trial. Neurology. 1997;49:351–357. [PubMed]
  129. Hohlfeld R, Wiendl H. The ups and downs of multiple sclerosis therapeutics. Ann Neurol. 2001;49:281–284. [PubMed]
  130. Paolillo A, Coles AJ, Molyneux PD, et al. Quantitative MRI in patients with secondary progressive MS treated with monoclonal antibody Campath 1H. Neurology. 1999;53:751–757. [PubMed]
  131. Jurewicz A, Biddison WE, Antel JP. MHC class I-restricted lysis of human oligodendrocytes by myelin basic protein peptide-specific CD8 T lymphocytes. J Immunol. 1998;160:3056–3059. [PubMed]
  132. Sobottka B, Harrer MD, Ziegler U, et al. Collateral bystander damage by myelin-directed CD8+ T cells causes axonal loss. Am J Pathol. 2009;175:1160–1166. [PMC free article] [PubMed]
  133. Cappellano G, Orilieri E, Comi C, et al. Variations of the perforin gene in patients with multiple sclerosis. Genes Immun. 2008;9:438–444. [PubMed]
  134. Matusevicius D, Kivisäkk P, Navikas V, et al. Interleukin-12 and perforin mRNA expression is augmented in blood mononuclear cells in multiple sclerosis. Scand J Immunol. 1998;47:582–590. [PubMed]
  135. Rubesa G, Podack ER, Sepcić J, et al. Increased perforin expression in multiple sclerosis patients during exacerbation of disease in peripheral blood lymphocytes. J Neuroimmunol. 1997;74:198–204. [PubMed]
  136. Kivisäkk P, Stawiarz L, Matusevicius D, et al. High numbers of perforin mRNA expressing CSF cells in multiple sclerosis patients with gadolinium-enhancing brain MRI lesions. Acta Neurol Scand. 1999;100:18–24. [PubMed]
  137. Deb C, Lafrance-Corey RG, Zoecklein L, et al. Demyelinated axons and motor function are protected by genetic deletion of perforin in a mouse model of multiple sclerosis. J Neuropathol Exp Neurol. 2009;68:1037–1048. [PMC free article] [PubMed]
  138. Derfuss T, Parikh K, Velhin S, et al. Contactin-2/TAG-1-directed autoimmunity is identified in multiple sclerosis patients and mediates gray matter pathology in animals. Proc Natl Acad Sci U S A. 2009;106:8302–8307. [PMC free article] [PubMed]
  139. Bartos A, Fialová L, Soukupová J, et al. Elevated intrathecal antibodies against the medium neuro-filament subunit in multiple sclerosis. J Neurol. 2007;254:20–25. [PubMed]
  140. Rawes JA, Calabrese VP, Khan OA, et al. Antibodies to the axolemma-enriched fraction in the cerebrospinal fluid and serum of patients with multiple sclerosis and other neurological diseases. Mult Scler. 1997;3:363–369. [PubMed]
  141. Mathey EK, Derfuss T, Storch MK, et al. Neurofascin as a novel target for autoantibody-mediated axonal injury. J Exp Med. 2007;204:2363–2372. [PMC free article] [PubMed]
  142. Moffett JR, Ross B, Arun P, et al. N-Acetylaspartate in the CNS: from neurodiagnostics to neurobiology. Prog Neurobiol. 2007;81:89–131. [PMC free article] [PubMed]
  143. Hulshagen L, Krysko O, Bottelbergs A, et al. Absence of functional peroxisomes from mouse CNS causes dysmyelination and axon degeneration. J Neurosci. 2008;28:4015–4027. [PubMed]
  144. Kassmann CM, Lappe-Siefke C, Baes M, et al. Axonal loss and neuroinflammation caused by peroxisome-deficient oligodendrocytes. Nat Genet. 2007;39:969–976. [PubMed]
  145. Dietz KC, Casaccia P. HDAC inhibitors and neuro-degeneration: at the edge between protection and damage. Pharmacol Res. 2010;62:11–17. [PMC free article] [PubMed]