Open access peer-reviewed chapter

Intrathecal Immunoglobulin Synthesis in MS—A Complete Reappraisal

Written By

Mickael Bonnan

Submitted: 12 October 2015 Reviewed: 18 March 2016 Published: 08 September 2016

DOI: 10.5772/63201

From the Edited Volume

Trending Topics in Multiple Sclerosis

Edited by Alina Gonzalez-Quevedo

Chapter metrics overview

3,249 Chapter Downloads

View Full Metrics

Abstract

Multiple sclerosis (MS) is characterized by the intrathecal synthesis (ITS) of immunoglobulins (Igs), which, although nonspecific, is the strongest biological marker. Since no specific target has been elucidated, this synthesis is considered to be disease‐irrelevant. We demonstrate that this synthesis provides pertinent information about the pathophysiological processes involved. Quantification of ITS is based on an approximation intrinsically underestimating its level and it remains constant in MS, albeit sometimes at a low level. B‐cell maturation seems to be initiated within the cervical lymph nodes and B‐cells traffic on both sides of the blood‐brain barrier by rounds of bidirectional traffic. During this process, they undergo somatic hypermutation, which is the hallmark of antigen‐driven antibody maturation, suggesting that most of the ITS is probably directed against as yet unknown targets. Alternatively, examining”non‐disease‐relevant” ITS in the light of meningeal tertiary lymphoid organs provides new insights into the pathophysiology of MS. Although no specific target has yet been identified in MS, recent developments in the search for targeted antigens point to non‐conventional antigens (posttranslationally modified proteins or oxidized products) of which a few are promising for future research.

Keywords

  • multiple sclerosis
  • meningeal inflammation
  • intrathecal synthesis
  • immunoglobulins
  • autoantibody
  • epitopes
  • antigens

1. Introduction

The normal central nervous system (CNS) is completely devoid of resident plasma cells, so finding infiltrating B‐cells in the CNS is rather exceptional. Infiltrating activated CD20+CD23+ B‐cells in brain parenchyma number to less than 0.1–1.5 cell/cm2 [13]. Therefore, no intrathecal immunoglobulin (Ig) synthesis (ITS) occurs in the basal state, and locally produced Igs testify to former or active CNS infection or inflammatory processes. Therefore, while it is easy to demonstrate high levels of ITS, quantifying low levels poses technical problems.

Although ITS is one of the most sensitive biological clues for MS, it was long considered to be nonspecific or a bystander of CNS inflammation since the latter mainly involves T‐cells and no specific antibody target has yet emerged. The recent success of anti‐CD20 therapy in relapsing and progressive MS sheds light on a more central role of non‐CNS B‐cells in the pathophysiology of MS. However, progressive MS pathophysiology is still poorly understood and CNS resident B‐cells may play an even greater role. Since locally synthesized Igs are the main B‐cell by‐products, ITS offers a good opportunity for studying resident B‐cells. The role of cerebrospinal fluid (CSF) B‐cells has not yet been elucidated and is probably diverse as follows: (1) cytotoxic effect of antibodies; (2) local antigen presentation to T‐cells; and (3) secretion of cytokines playing a complex regulating role, including a possible Ig‐unrelated CNS cytotoxicity. Therefore, CNS‐trapped B‐cells not only synthesize oligoclonal bands (OCBs) but might also play a pivotal role in the slow‐burning CNS inflammation.

Advertisement

2. Quantitative measures of CSF IgG

2.1. Pitfalls in assessing ITS level

Figure 1.

CSF IgG passively diffused from blood to CSF in normal population. Left panel (A). Plot of CSF/serum quotients with hyperbolic function of quotient ratios (“'Reibergram”). Reference range defined by QLim : Qmean±2SD or ±3SD involving, respectively, 96 and 99% of the normal population. Dotted line is the upper normal limit (>0.7) of IgG index, intersecting the QLim curve at two points. Right panel (B). Probability curve of basal QIgG for a given QAlb. The maximum probability is obtained for Qmean. Points are obtained from a simulated healthy population.

Synthesis of Igs does not occur in the normal CNS and the tiny Ig concentration measured in normal CSF reflects a low‐rate passive diffusion through the blood‐brain barrier (BBB) to CSF. It was long thought that although virtually all the molecules may diffuse from serum to CSF, BBB permeability positively correlated with the molecular weight [4]. For example, the ratio decreases from 1:205 with albumin (65 kDa) to 1:440 with IgG (150 kDa) and 1:900 with IgM (970 kDa) [5]. Moreover, the permeability of the BBB commonly increases during CNS pathologies, leading to an increase in CSF concentrations of blood‐borne proteins and Igs. As a consequence, the level of intrathecally synthesized Igs related to CNS inflammation is obscured by the passively diffused CSF Igs concentration and requires a mathematical approach that takes into account the permeability and blood concentration of the targeted molecules. The albumin quotient (or ratio), QAlb = [AlbCSF]/[Albserum], is indicative of BBB dysfunction and increases with its permeability. It is also influenced by the patient's age and underlying CNS pathologies. In the basal state, which is devoid of intrathecal IgG synthesis, the ratio [IgGCSF]/[IgGserum] is proportional to QAlb.

The daily IgG synthesis rate can be assessed by Tourtellotte's formula (median value 29 mg/day), but inconsistent values are obtained at the individual level owing to assumptions [6]. The linear approach where the IgG index = QIgG/QAlb (normal values <0.7) does not take into account either normal QIgG variance nor nonlinear correlations with QAlb, leading to an approximation around the limit (Figure 1, left panel). Moreover, since QAlb increases with age, the IgG index is thought to decrease mechanically without any change in ITS [7]. Data are best fitted by an empiric hyperbolic function (the “Reibergram”) [8] whose constant parameters were fitted with a large dataset of 4154 control patients [9]. From a theoretical point of view, the hyperbolic function is the application of Fick's laws of diffusion applied to albumin and Igs [9]. For a given QAlb in a population of normal patients, the distribution of QIgG follows a normal law around the mean curve as Qmean=f(QAlb) (Figure 1, right panel) [9]. With intrathecal synthesis (ITS), the IgG concentration in the CSF is the sum of IgG passively diffused from the blood and synthesized intrathecally

Q I g G =   ( [ I g G C S F _ p a s s i v e ] + [ I g G C S F _ L o c ] )   /   [ I g G s e r u m ]   = Q I g G _ b a s a l + Q I g G _ L o c , E1

where QIgG_basal is the QIgG of the same patient before the onset of ITS. In clinical practice, only QIgG is directly available but not QIgG_basal, so the exact IgGCSF_Loc concentration can only be approximated by using Reiber's discrimination curve. The upper limit of the reference range, QLim, is usually set arbitrarily as Qmean+3SD and involves >99% of the normal population. By applying this definition, intrathecal IgG synthesis is considered to be present when QIgG> QLim. The major drawback of this reference range is the loss of sensitivity for cases displaying a low level of ITS (QLim> QIgG> QIgG_basal). In common practice, demonstrating ITS in such cases requires a CSF‐restricted OCBs positivity. As expected, restricting the reference range to QLim+2SD instead of +3SD increases the percentage of abnormal QIgG in MS cohorts by 6–10% for IgG and up to 20% for IgM, which increases the risk of false positivity to 4% [7].

The true amount of intrathecally (or locally) synthesized IgG should be calculated as follows:

I g G L o c ( m g / L ) =   ( Q I g G Q I g G _ b a s a l )   x   [ I g G s e r u m ] . E2

However, since QIgG_basal is unavailable, it can be replaced by either QLim or Qmean. As previously demonstrated, replacing QIgG_basal by QLim confers maximal specificity in single patient studies but with the drawback of an unavoidable underestimation of ITS. The range of IgGLoc underestimation may vary widely from 1 to 50 mg/L according to QAlb (unpublished results). In cohort studies, QIgG_basal is advantageously replaced by Qmean, which provides a closer estimation of the exact IgGLoc [7].

Lastly, inter‐assay variability may directly impact QIgG or the IgG index. For example, a 10% decrease in serum IgG directly means a 10% increase in IgG index, and 10% variations of the IgG index from day to day are commonly reported [7, 10]. As a consequence, minor fluctuations in IgG levels may be translated into normal or abnormal QIgG results, although the intrathecal IgG synthesis rate is not really impacted. A final pitfall of ITS assessment relates to the properties of CNS‐targeting antibodies themselves, which are capable of brain adsorption that can potentially abolish low levels of specific antibodies that are synthesized locally or spill over to the CSF [11].

2.2. Predicted changes in ITS measures in response to treatments

Using formulas and normal values obtained from the literature, we simulated results of a cohort with tunable ITS level (unpublished results). This model provides the advantage of being able to compare the calculated (approximated) IgGLoc with the fixed IgGLoc.

IgGLoc estimation based on Qmean fitted well with the exact IgGLoc, even in small cohorts and for small ITS (<1 mg/L). On the other hand, individual or cohort estimations of IgGLoc based on QLim were strongly biased in a range dependent on QAlb.

Figure 2.

Effect of plasma exchange on QIgG. Simulated population with ITS +5 mg/L. QIgG values increase to abnormal range whereas ITS level remains unchanged.

Plasma exchange depletes both serum IgG and IgG passively transferred through the BBB. Therefore, assuming that ITS remains constant during the procedure, the contrast in locally synthesized and passively diffused IgG in CSF is dramatically tuned by plasma exchange (Figure 2). For example, after a 90% decrease in [IgGserum], IgG in CSF originates almost entirely from local synthesis. The IgG index increases whereas the precision of the IgGLoc calculation (based on Qmean) remains unchanged.

For a given level of ITS, a decrease in BBB permeability decreases QIgG_basal in a nonlinear response whereas QLoc remains constant. Therefore, QIgG and the IgG index may shift to abnormal values and IgGLoc(QLim) increases incorrectly, whereas IgGLoc(Qmean) remains constant.

As a consequence, monitoring of the ITS level should be based on the unbiased IgGLoc(Qmean).

Advertisement

3. Qualitative measures of CSF IgG—method and limitations of oligoclonal band analysis

3.1. Oligoclonal bands

CSF OCB analysis is based on an isoelectric focalization (IEF) technique fractionating Ig into multiple bands according to their respective pI. Structurally diverse antibodies sharing the same pI may cofocus in a single band [12]. Studying IgM OCBs is harder owing to the dissociation of pentamers before the F fragment and the arbitrary reassociation of the separated monomers [13].

Moreover, owing to differential posttranslational modifications (PTMs) (glycosylation or amino acid modifications), each particular IgG clone may display several pKs and may run in multiple bands in IEF conditions [14, 15]. Monoclonal antibodies occurring naturally (monoclonal gammopathy) or commercially available ones are resolved in multiple bands on IEF gel runs (ladder pattern) [1517]. In a series of 20 myelomas, IEF identified 5–10 OCB in serum [18]. Commercial monoclonal antibodies produced by hybridomas (monoclonal mouse antibody) or monoclonal IgG produced in vivo by plasma cell tumors (multiple myeloma) are highly heterogeneous and present a clone‐specific profile of glycosylation [17, 19]. Lastly, each OCB is a visual optical interpretation of a local contrast of color density differentiating OCB from the surrounding polyclonal background. Apart from the relative subjectivity of the technique, faint and ambiguous OCB may be obscured by a dense polyclonal background. Capillary IEF seems to increase sensitivity and demonstrated OCB in negative patients with classical IEF [20]. Therefore, immunoblotting against known antigens, for example, in association with viral encephalitis, unmasks specific OCB in cases failing to demonstrate any OCB with conventional IEF [21]. In MS patients explored for specific antibody anti‐viral synthesis, OCB against multiple viruses may occur in the same migrating zone on IEF [22] and CSF OCB against myelin basic protein (MBP) or measles obtained with immunoblot detection does not comigrate with OCB obtained by IEF [23].

There may be several consequences of this as follows: (1) each OCB may be composed of multiple distinct IgG, which may share by chance the same isoelectric point [15]; (2) multiple OCB in MS may derive from one or a small number of cell clones [17]; (3) OCB count cannot predict the number or variety of B‐cell clones; and (4) clonally expanded B‐cell clones may precede OCB detection, thus reflecting the difficulty to detect very low levels of ITS.

ITS in MS is characterized by numerous unambiguous OCBs (≥10 OCB) that are highly specific of MS [24]. Monoclonal bands are exceptional and should be considered carefully. Repeated LP in these atypical cases demonstrates a broadening of ITS or a different diagnosis [25]. Owing to their lack of specificity [25], ≥2 OCB are required in most studies to define an oligoclonal pattern with optimal specificity.

Absence of OCB (type 1 pattern) is observed in less than 10% in recent studies, whereas most MS patients display the type 2 pattern and only some have type 3 (Figure 3). Among OCB+ patients, patterns 2 and 3 are observed in about 90 and 10% in relapsing‐remitting (RR) and secondary progressive (SP) patients, respectively, whereas the distribution is 40 and 60% in primary progressive (PP) patients [27], suggesting that pattern responses may be linked to pathophysiology.

Figure 3.

Types of isoelectric focusing patterns on agarose gels [26]: Type 1, no OCB in CSF or serum (normal pattern). Type 2, OCB restricted to CSF, absent in serum. Indicates low‐level ITS. Typical pattern in MS. Type 3, Identical OCB in both serum and CSF with extra bands in CSF. Pattern seen during systemic synthesis associated with intrathecal synthesis. Seen in MS. Type 4, OCBs in CSF mirror those in serum. Indicates a systemic IgG synthesis and passive transfer of OCB from blood to CSF, without any local synthesis. From a purely theoretical point of view, a low local synthesis with similar B‐cell clones cannot be completely deciphered from this situation. Type 5, Ladder‐type identical OCB in both serum and CSF typically associated with monoclonal IgG proteins. Peripheral IgG synthesis without local synthesis.

3.2. Absence of OCB in MS patients is a technical limitation

OCB are almost always present when QIgG> QLim, but are also commonly present when QIgG QLim [26, 28, 29]. Nonetheless, about 5–10% of MS patients fail to demonstrate any OCB or an elevated IgG index. Nevertheless, the question remains whether ITS is really absent from such patients’ CSF. Our short review argues for a probable faint ITS in the rare ‘CSF‐negative’ patients, making ITS the most valuable marker of MS to date.

A high CSF IgA synthesis has been demonstrated in a few patients lacking the classical IgG synthesis [30, 31]. Free light chain (FLC) sensitivity seems to be near 100% but the (expected) specificity lower than OCB makes FLC less useful for routine clinical purposes [32, 33]. Oligoclonal κ‐FLCs are detected in about 50% of MS patients without OCB [34, 35]. Isoelectric focusing with affinity blotting against known antigens overcomes the limitation induced by the background and unmasks specific ITS. OCBs against the neurotropic MRZ viruses, measles, rubella, and varicella zoster virus (VZV), which are commonly observed in MS for reasons that remain unclear, were present in the CSF of 72% of MS patients who otherwise failed to demonstrate OCB with IEF [36].

The specific antibody index (AI) is calculated by using the ratio of specific antibodies QSpec=[SpecCSF]/[Specserum] in the formula, AI = QSpec/QIgG, where AI values >1.3–1.5 indicate the presence of ITS. An MRZ pattern, which is defined as an elevated AI against ≥2 neurotropic viruses, is observed in up to 90% of MS patients [37, 38]. However, the AI is not usually assessed in patients with QIgG QLim and negative OCB, because they are thought to be MRZ‐negative since AI correlates with QIgG [29]. When multiple AIs (VZV, herpes simplex virus (HSV), cytomegalovirus (CMV), measles, rubella, and Borrelia) were systematically assessed in MS patients without apparent ITS (QIgG QLim and negative OCB), all of them showed ≥1 MRZ reactivity and up to 47% of patients had OCB against ≥1 MRZ [29, 36, 3941]. Interestingly, AI results are not completely congruent with those obtained by IEF with affinity blotting, thus increasing the prevalence of ITS detected by ≥1 technique to 64% in the “CSF‐negative” subgroup of MS patients [36]. Since the MRZ reaction is common but not exclusive and because reactions against many other viruses have been confirmed in MS (see below), a larger antigenic test panel might improve the frequency of ITS detection [29], for example, by the systematic use of antigen array [42] and the systematic assessment of IgM.

Studies combining all currently available techniques should be undertaken in MS patients, especially in those with “negative CSF”, in order to establish the true prevalence of ITS in MS, especially in patients of non‐Caucasian descent who are thought to harbor a lower CSF positivity [43, 44]. Future studies could combine quantitative techniques (IgGLoc, FLC, AI against MRZ) and highly sensitive qualitative techniques (OCB) until the discovery of putative antigenic targets such as anti‐CCP (cyclic citrullinated peptide) in rheumatoid arthritis. Criteria for standardizing intrathecal Ig synthesis should be based on the simultaneous normalization of all the tests. In view of the fluctuations of ITS in individual patients (up to 30% of IgGLoc [45]), the action of a drug upon ITS should be statistically demonstrated in groups. To be able to demonstrate a successful intrathecal reset, a null Ig synthesis should be confirmed by using several techniques together.

Advertisement

4. Intrathecal Ig synthesis is a robust and predictive marker in MS

4.1. Factors influencing ITS level

About 95% of patients display OCB but a quantifiable IgG synthesis only occurs in about 70% of them. Several studies have shown that the level of ITS is highly variable from one patient to another, which suggests a genetic determinism. Females are more prone to OCB positivity (92% vs 84%) and to a higher IgG index (1.32 ± 0.92 vs 1.03 ± 0.54) [46, 47]. An ethnic influence on IgG index level and OCB status has also been confirmed by several studies. Black‐ascending patients are more prone to a higher IgG index, a higher rate of IgG synthesis, and OCB positivity [48, 49]. The IgG index level is still higher in Afro‐Americans after adjustment for OCB status [49]. The association of female sex to higher IgG index was also confirmed in this group [49].

Prevalence of abnormal OCB status and IgGLoc is lower in Asian patients than in Caucasian patients [5052]. In Asian patients having the strongest genetic risk factors (DRB1*1501), the prevalence of OCB positivity remains lower than in Caucasians (<72% vs 97%) [53, 54]. Moreover, irrespective of genetic background, extreme latitudes may exert a positive influence on OCB status whereas tropical latitudes exert a negative influence [53, 55].

Several studies have confirmed a strong association between OCB status and human leukocyte antigen (HLA). The DRB1*15 allele confers a higher risk of OCB positivity whereas the genotypes DRB1*04:04/*04:05 or DRB1*03:01/*04:01 are associated with OCB negativity [56, 57]. Interestingly, DRB1*1501 also increases the prevalence of abnormal IgG and IgA indexes but not of the IgM index [54]. In Asian patients, even though these determinants are conserved, OCB status is also driven by a strong interaction with latitude [53].

Genetic markers of γ chains (GMs) display 18 serologic specificities and all of the variants but two are expressed in the Fc region of γ chains. Some of these variations have been shown to strongly influence antibody‐dependent cell‐mediated cytotoxicity (ADCC) and complement‐dependent cytotoxicity (CDC) [58]. In an animal model of rheumatoid arthritis, the recognition of collagen‐II epitopes by VH chains strongly depended on GM haplotypes [59]. Interestingly, GM haplotypes are in almost total linkage disequilibrium among world populations and are therefore typical of geographic origin unless there is a genetic admixture [60]. Gm21* haplotypes were associated with high ITS in a Caucasian population although Gm5*;3 haplotypes are associated with a low level of ITS96, 97. Studies are required concerning non‐Caucasian (African and Asian descent) haplotypes. Gm haplotypes are neither correlated with serum IgG level nor with the risk of MS [61, 62]. The causes of interaction between GM allotypes and ITS level remain elusive and the relationship between allotypes and CNS FcRn function (which enhances IgG clearance from CSF via Fc interaction) is unknown. Other non‐HLA unknown genes also influence OCB status [56, 63].

Healthy siblings of MS patients display a hyperimmune condition termed “MS immunopathic trait” that is characterized by (1) one or more CSF OCB, (2) an exaggerated response to a variety of viral antigens, and (3) an increased BBB permeability [64, 65]. CSF OCB were found in up to 19% of MS siblings in contrast to 4% of an unrelated control population [66, 67], some of them also displaying an elevated IgG index [66] and up to half of these siblings having both OCB‐ and measles‐specific IgG local synthesis [64, 67]. It is not clear whether ITS observed in these cases reflects a presymptomatic MS in high‐risk patients, elite non‐progressors, or an unrelated predisposition to ITS.

4.2. ITS is robust over time

ITS occurs as a very early disease event. In pediatric cohorts with very early onset MS (before 6 years), only 8% showed IgG OCB, a figure increasing to 90% with 69% of intrathecal production in two early onset cohorts (<15 y) and even more after a further relapse [68, 69]. In adults, the proportion of OCB positivity tends to appear at re‐examination [70] or increases as the disease progresses (RR = 325/360, SP: 25/25, and PP: 39/39) [27]. In a longitudinal study of CSF from 19 patients taken at multiple time points up to 12 years (mean 8 years), OCB never abated with time and demonstrated a robust pattern [71]. Most of the changes concerned band intensity and the acquisition of a new band [25, 72], although band loss was sometimes described in earlier reports [25, 71]. The same stability of OCB over time was observed with IgG, IgM, and IgA [13, 71, 73].

When focusing on the clonal repertoire of CSF Ig, the highest number of clones was found in patients with the longest disease duration, suggesting a continuous clonal expansion over time [74]. However, the low number of patients included cannot totally rule out disease heterogeneity [74]. In a longitudinal study of CSF B‐cells in two MS patients, clonal rearrangements on VH (VH3 and VH4), D segment, JH, and HCDR3 sequences were conserved at two time points 1 and 4 years apart [75]. The antibody index of phagotopes from the entire panel of clones extracted from a single patient was stable at 2 years [76]. The peptidic targets of the OCB IgG are constant over time [72] and the activity index (AI) against infectious agents also remained stable over several years [77, 78]. These findings are in line with a slightly delayed oligoclonal immune response immediately after the onset of MS. However, once initiated, CSF IgG secretion persists over time indefinitely and is little altered qualitatively at the clonal level [71]. Other CSF parameters such as albumin index, IgG, and IgM index remained essentially the same in the clinically isolated syndrome (CIS), RR‐MS, and progressive MS groups [27, 79].

A few exceptions should be mentioned as follows: (1) IgM OCBs behave differently, if absent at onset, no further IgM OCBs appear [73]; when present, they tend to disappear after a few years (RR: 109/360, SP: 15/25, PP: 0/39) [27]. When oligoclonal IgMs are present, the disease lasts for less than 5 years in 90% of patients, but when IgMs are absent, it lasts for more than 5 years in 60% of patients. (2) Anti‐MBPs in CSF are high during relapses in about 90% of patients but vanish during the 6 weeks following onset, relapse treatment appearing to precipitate this event [80, 81].

Each patient has a private and unique CSF OCB pattern, the so‐called OCB fingerprint [82, 83]. The pattern persists even though the total CSF IgG is significantly reduced, for example, by steroids [83]. The CNS IgG synthesis rate remains roughly stable over several months [6]. When steroids are used to reduce the IgG index, pretreatment values are recovered in a few weeks or months [83]. Intrathecal steroids rarely obliterate OCB for more than 1 month [71], but this modification can be corrected when equal amounts of CSF Ig are applied in IEF.

Unfortunately, OCBs provide basic information and the future development of microarrays may provide more precise insight into the intimate evolution of clonality. For example, the involution of the antibody response against heat shock protein (HSP) in serum is a central modification in the immune response during the transition from RR to the progressive form [84], but the outcome of this immune signature in CSF is unknown.

The persistence of OCB suggests a local IgG production by long‐lived plasma cells residing inside the CNS, suggesting that the B‐cell fostering properties of the CNS deserve consideration [85]. On the other hand, minor changes in OCB pattern are in accordance with competition between plasma cells for a limited number of survival niches, as observed in the bone marrow. It seems unlikely that such a discrepancy between brain volume and the small number of CNS plasma cells can be explained only by the B‐cell fostering properties of the CNS. Moreover, the intrathecal IgG synthesis rate is roughly stable over time, even though persistent local inflammation would be expected to induce a gradual increase in synthesis rate. These data suggest that the number of CNS niches is limited and that they are quickly saturated. Therefore, the putative role of CNS tertiary lymphoid organs (TLOs) deserves further consideration.

4.3. None of the available MS drugs deplete intrathecal Ig synthesis

In a complete review of the literature, we demonstrated that none of the available (approved or experimental) treatment proved to abate intrathecal Ig synthesis (review in reference [86]). A null or minimal (<20%) impact on intrathecal IgG synthesis was observed after the administration of azathioprine, β‐interferons, cytarabine (intravenous or intrathecal), lomustine, 5‐fluorouracil, CNS irradiation, mitoxantrone, methotrexate, cyclophosphamide, cyclosporin A, cladribine, alemtuzumab, daclizumab, fingolimod, or stem cell transplantation following myeloablation.

Steroid infusion decreases blood Ig levels in a few days [87]. However, high‐dose steroid infusions have little effect on IgG index and OCB number [8890], irrespective of the dosage or the mode of administration (intramuscular, intravenous, and intrathecal) [91]. Even if steroids can transiently lower the IgG index in most but not all patients, the decrease in the range of CSF IgG synthesis is low [90, 92]. The mean antibody reactivity and the mean number of targeted antigens in CSF are decreased in the 2 months following steroids [93].

Rituximab depletes CSF B‐cells but fails to significantly decrease intrathecal IgG synthesis (<-20%) [45, 94], possibly because ITS may partly depend on plasma cells (CD20-), which are constitutively resistant. This failure to lower intrathecal IgG secretion was predictable from the absence of the effect of blood‐infused rituximab upon serum IgG and IgA levels, contrary to a minor effect upon IgM levels [45, 95]. No data are available on intrathecal IgM synthesis after rituximab treatment.

Given the low diffusion of rituximab to CSF (<0.2%) and the growing body of evidence demonstrating the safe use of intrathecally infused rituximab, a rationale to infuse intrathecal rituximab in progressive MS recently emerged (review in reference [96]). Data obtained from a single patient receiving intrathecal rituximab (10 mg per month for 2 months) showed a major effect upon CSF cytokine levels although intrathecal IgG synthesis was unchanged [97, 98] (unpublished).

An unexpected partial repression of intrathecal IgG synthesis was obtained with natalizumab treatment in some series [99101] but results were null or lower in two others [102, 103]. These findings need to be replicated in another large cohort. Three non‐mutually exclusive explanations may be put forward. First, α4β1‐integrin is expressed by B‐cells (CD19+ and CD138+) at higher levels than CD3+ T‐cells, and natalizumab impedes B‐cell trafficking to the brain and renewal of the CNS plasmablast pool. Second, apart from cytokines, T‐cells also play a supportive survival role in the survival niches of plasma cells [104]. Third, natalizumab inhibits the CNS migration of dendritic cells, which in turn may affect the maintenance of CNS lymphoid tissue [105], and dendritic cell counts in MS brains were lower than expected [106]. Of note, a significant decrease in IgM (≈-45%) (and less significantly in IgG1≈-18 to 36%) plasma levels also occurs during natalizumab treatment but is not correlated with treatment duration, suggesting that it effectively perturbs the IgG synthesis pathway [103, 107]. Future experiments should examine the kinetics of intrathecal IgG secretion after withdrawal of natalizumab. Data obtained from a single patient devoid of OCB under natalizumab and discontinuing treatment for progressive multifocal leukoencephalopathy (PML) showed that OCB returned shortly with a slightly modified pattern [101], whereas a sustained negativation was observed at 6 months in two other patients [99]. However, in a series of 23 patients, no increase of ITS or OCB was observed at 14 months after natalizumab discontinuation [108], suggesting that the impact of natalizumab on ITS is probably low.

A single patient treated by EBV‐specific adoptive immunotherapy (autologous CD8+ T‐cells activated against EBV) regained a normal level of IgG index at month 4 [109]. This single case needs to be replicated.

4.4. Prognostic value of ITS

Although the prognostic value of OCB positivity is not consensual, ITS is consistently associated with a worse outcome. In CIS patients, none of the IgG, IgA, or IgM indexes are predictive of clinical conversion to MS [110]. On the other hand, OCB positivity in CIS increases the risk of having a second attack irrespective of magnetic resonance imaging (MRI) results [111] and the number of OCB is an independent risk factor for relapses [112], whereas their absence is associated with a lower clinical conversion at 5 years and with a better clinical outcome [113]. Interestingly, double OCB positivity (IgG and IgM) is associated with an odds ratio (OR) of second relapses higher than fulfilling of Barkhof's criteria (OR 55.7 vs 3.5) [114]. The MRZ pattern is also an independent risk factor of clinical conversion [115] and all CIS patients displaying OCB targeting lipid antigens relapse in the first year [113].

In clinically defined MS patients, the IgG synthesis rate correlates with MRI burden and the IgG1 index correlates with expanded disability status scale (EDSS). Free light chains in CSF are predictive of impairment [116]. CSF IgM and IgM OCB are associated with a poorer clinical outcome [73, 117] and a lower brain volume [118, 119]. IgM OCBs reacting against myelin lipids are associated with a higher relapse rate, lower efficiency of IFNβ, and a faster EDSS increase [120, 121]. Time and probability to reach EDSS 3 and 4 are strongly correlated with the presence of IgM in CSF [122, 123]. The IgM index in CSF at onset is strongly correlated with a higher last follow‐up EDSS [117, 124]. The time to conversion to the secondary progressive phase is shorter in the presence of IgM BOC or an elevated IgM index [73, 123, 124]. These attractive findings concerning IgM were established in a single group of patients. Stauch et al. [13] did not find any influence of IgM OCB on the evolution of MS in a pediatric group and raised some technical concerns about the IgM tests used previously, which may invalidate the results. Another group claimed negative results but they also obtained a shorter, albeit nonsignificant, time to relapse after CIS in OCB‐IgM+ patients [125]. In a large retrospective study, disease severity appeared to be identical between OCB‐positive and ‐negative patients [126], whereas in a study including 1800 MS patients, the number of OCB was considered to correlate with the course of the disease [127]. The mean number of OCB in the group of benign patients was 2.9 ± 3.6 compared to 5.7 ± 4.9 in the severe group (p < 0.06) [127]. Therefore, the evidence points to a poorer clinical prognosis in MS patients in association with ITS. On the other hand, OCB negativity correlates with a better outcome in many studies [70, 127131], even after demographic adjustment [132].

Advertisement

5. Intrathecal Ig synthesis in animal models—technical limitations

IgG synthesis level in CSF is highly correlated with CD19+CD138+ plasmablast levels [133], but although ITS is commonly thought to be associated with CSF floating cells, this minute cell number may account for less than 0.1% of the whole ITS, meaning that the bulk of IgG synthesis is provided by resident IgG‐secreting cells, residing either in the meninges or in the perivascular areas [134]. In experimental allergic encephalomyelitis (EAE) models, the parenchymal level of B‐cell infiltration increases from null to 134 B‐cell/cm2 [135] and the whole number of infiltrating B‐cells is in the range of 103–105 cells per mouse or rat brain [136, 137]. In MS, B‐cells remain a minor proportion of the infiltrating lymphocytes (<5%), are virtually absent from the parenchyma, and are mostly observed around the small veins and meninges (4.6–6 cells/mm2)[1, 138]. Plasma cells are occasionally found in demyelinated areas. No data are available regarding a possible role of meningeal B‐cell aggregates in ITS.

IT Ig synthesis in animal models has received little attention since the very small volume of CSF does not facilitate sampling. Although older experiments based on low‐sensitivity techniques failed to demonstrate any ITS during EAE, optimal techniques (IEF followed by anti‐IgG staining) have subsequently confirmed the local synthesis of OCB and/or an elevated IgG index [139142]. In viral models of demyelination (i.e., measles or JHM strain of coronavirus infection), a mirror pattern of OCB is common but local OCBs are always revealed by immunoblot against viral antigens [21, 143]. We are not aware of any EAE experiments aimed at demonstrating the nonspecific pattern of ITS observed in MS, especially the animal counterpart of the human MRZ pattern using common animal viruses or vaccines. From a theoretical point of view, animals receiving intrathecal vaccination with foreign antigens such as albumin are able to mount a strong intrathecal response [144146]. In a unique case challenged intrathecally with ovalbumin (OVA), an increase in AI‐herpes was also obtained apart from the expected increase in AI‐ovalbumin, suggesting that animal models are promising for future studies of nonspecific ITS [145].

Advertisement

6. Non‐IgG ITS

6.1. General background

The IgG class accounts for the bulk of locally synthesized Igs in MS and most knowledge to date concerns this class. All Ig classes may give rise to ITS. IgG, IgM, and IgA are, respectively, present in 100, 20–36, and 6–9% of MS patients [69, 78]. The relative absence of IgE is commonly interpreted as a clue to type I immunity [147].

6.2. IgM chains

Intrathecal IgM synthesis is always higher in MS than in controls [110]. IgM OCB may occur in about 20–75% of MS patients [78, 113, 118] and in 74% of CIS [125]. CD19+CD5+ lymphocytes are the predominant population of B‐cells in CSF and are characterized by the T‐cell independent secretion of IgM natural antibodies directed against phylogenetically conserved structures that target non‐proteic epitopes. In a series of 53 CIS/relapsing‐remitting multiple sclerosis (RRMS), 46 of them had oligoclonal IgM‐recognizing lipid antigens, 30 targeted phosphatidylcholine (PC) alone, seven targeted PC and had additional reactivity against other myelin lipids (phosphatidylethanolamine, phosphatidylinositol, and sphingomyelin), and nine targeted myelin glycolipids, mostly sphingomyelin [113]. IgM OCB against lipids may be restricted to the RR and SP phases since they were not observed in PP patients [27]. Pentameric IgM strongly activates the complement cascade and is involved in the lesions of plaques (pattern II).

6.3. IgA

The IgA index is elevated in at least 9% of MS patients [54, 78, 148] and the mean intrathecal IgA index is modestly but significantly higher in patients than in controls [110]. Local synthesis of IgA may be the only clue to local Ig synthesis in some MS patients devoid of local synthesis of IgG [31]. About a quarter of Ig‐secreting cells are IgA in choroid plexuses [149]. IgA cells are also common in plaques (3–9%) and demonstrate a high clonality (up to 80%) [149]. IgAs are mainly mucosal secretory antibodies but could be a major component of CNS immune responses in certain viral infections [148, 150]. OCB IgAs were reported to have a low frequency (2/33) in the earlier CSF studies [71, 150]. In a post‐mortem analysis of plaques extracted from four MS brains, IgA+ plasma cells were recovered in 18/24 plaques and sometimes predominated on Ig+ plasma cells [148]. Clonality assessment in the plaques of two patients demonstrated a low diversity of clones and a clonal restriction confined to plaques [148]. The somatic mutations were highly concentrated in the complementary determining region (CDR) and FR3, which are considered critical for antigenic specificities, suggesting an antigen‐driven maturation of affinity as observed for IgG. The locally synthesized IgA were demonstrated to target injured axons [148].

6.4. Free light chains

Light chains are normally synthesized more than heavy chains, with an FLCκ/λ ratio of 2:1 in normal blood. Serum circulating levels of FLC are about 1000‐fold lower than Ig levels owing to rapid renal clearance (serum half‐life of 2–6 h), whereas the rate of clearance from CSF is similar with Ig [151]. Therefore, the level of κFLC in MS is equal or higher in CSF than in serum. Although the absolute value of CSF FLC is variable among studies, the FLC level is always higher in MS than in controls, and the κFLC/protein ratio is even higher [152]. The κFLC index is more sensitive (Se 95%, Sp 91%) than the IgG index and OCB [153]. Since elevated CSF FLC is predictive of impairment [116], it has even been proposed as a therapeutic target [47].

Advertisement

7. Clues suggesting an antigen‐driven maturation of the intrathecal clonal population of B‐cells

The IgG1 subclass is significantly elevated in most MS CSF although some authors recovered rare higher CSF IgG2 or IgG3 indexes [110, 154]. The Ig repertoire uses 51 functional germline genes and the normal repertoire of naïve peripheral blood CD19+ cells closely approximates the frequency of the VH family gene segments within the germline. On the other hand, over‐representation of a single VH family is common in MS plaques and CSF but not in blood [155]. The VH4 family is observed in more than 35–95% of CD19+ or CD138+ cells. However, the VH1 family, which represents 20% of the functional germline, is nearly absent from CSF in MS [156, 157]. As observed for IgG, the VH4 germline predominates in CSF IgM [158]. This bias precedes the onset of OCB [155] and is typical of MS whereas different biases are observed in other CNS disorders [155]. The clonal expansion of a single ancestor gene was robustly demonstrated by the analysis of clonal diversification from an ancestor gene accumulating substitutions [156]. In CIS patients, the presence of a VH2‐VH4 CSF bias either in CD19 or in CD138 cells, which is observed in 7/10 patients, was highly correlated with a clinical conversion to MS in the following 6 months. By contrast, none of the CIS patients without the repertoire bias had developed MS at 2 years of follow‐up.

Somatic hypermutation (SHM) drives the maturation of the variable regions of the Ig genes under the control of activation‐induced cytidine deaminase (AID), with a preference to the hotspot regions (RGYW/WRCY motifs) of the CDR. The antigen‐driven affinity maturation process positively selects replacement mutations occurring in the CDR, which are in contact with the antigen, whereas silent mutations predominate in the framework regions. Therefore, the R/S ratio, which is lower than 2.9 before antigen‐driven maturation, is expected to be higher in CDR and the same or lower in the framework regions. In MS, the R/S ratio in the CDR of Ig genes was unchanged in blood B‐cells but elevated in CSF B‐cells [149, 158]. This ratio is elevated for both IgG‐ and IgM‐producing CSF B‐cells and occurs to the same extent in CSF, whereas the blood IgM ratio is about half of the IgG ratio [158]. The extent of SHM is in the expected range in CSF naïve B‐cells, whereas its level increases in switched memory B‐cells and plasma cells [159].

It is widely admitted that AID expression is a stage‐specific hallmark of germinal center B‐cells undergoing SHM and gene conversion. However, an aberrant expression of AID was demonstrated in CSF B‐cells producing IgG and IgM [158].

A signature score of hot/cold spots of higher/lower frequency replacement was established with six key positions, mostly situated in the CDR [160]. The signature score was 4.5 in the CSF of non‐MS patients, 2.0 in the blood of MS patients, and 10.9 ± 2.0 (7.6–11.9) in the CSF of MS patients (cutoff predictive of clinically defined MS ≥6.8). This technique was applied to B‐cells extracted from the autopsied brains of four MS patients. The scores ranged from 10 to 14.5, so B‐cells from plaques and CSF share the bias [161]. Unfortunately, these bench results cannot be translated into common practice. Various clones displaying the same amino acid sequences but encoded by different nucleotides reinforce the hypothesis of antigen‐driven affinity maturation [158].

In summary, B‐cells in CSF demonstrate the cardinal features of an antigen‐driven humoral immune response: clonal expansion and somatically hypermutated VH family sequences [155]. This suggests that these lineage cells were expanded by antigen and have undergone a germinal center reaction [162]. Moreover, there is evidence that the maturation process takes place at least partly in the CNS compartment [163].

Advertisement

8. Maturating B‐cells undergo bidirectional exchanges across the BBB

Figure 4.

Schematic B‐cell lineage tree and putative migration routes. Lineage founding B‐cell is postulated in the CLN (green), which drains antigens and B‐cells coming from the CNS. Immunoglobulins are subjected to somatic hypermutation (SHM) and proliferation in cervical lymph nodes (CLN). Migrating B‐cell clones are sometimes encountered in blood on their way to the CNS. B‐cell proliferation occurs both in CLN and in CNS. Evidence is growing that SHM may also occur inside the intrathecal compartment, possibly in meningeal tertiary lymphoid organs (TLOs). In such a model, rounds of B‐cell maturation and proliferation may occur in a recursive way both in CNS and in CLN [14, 149]. Long‐lived plasma cell (PC) differentiation occurs in the different compartments.

Authors are often reluctant to localize the SHM process inside the CNS and prefer models that posit peripheral antigenic stimulation followed by CNS migration, although evidence is accumulating in favor of a local maturation process. High‐throughput sequencing techniques allow the analysis of clonal B‐cell populations on both sides of the BBB. A common finding is the demonstration of IgG cell lineages either restricted to one compartment (CNS, blood, and lymph node) or overlapping multiple compartments. In a study pairing CNS and cervical lymph node (CLN) B‐cells, about 6–15% of the IgG B‐cell sequences in plaques were recovered in the CLN [149]. Cell lineage analysis demonstrated that most of the founder cells originated in the CLN [149]. A tentative interpretation is that antigen‐driven affinity maturation of B‐cells takes place in the CLN, which drains CNS antigens [164], then B‐cells migrate to colonies populating the CNS and continue to traffic between the CNS and the periphery, notwithstanding the possibility that a bidirectional traffic occurs in association with clonal expansion in both compartments (Figure 4) [149]. Clonal populations of CSF IgM‐ and IgG‐secreting B‐cells do not overlap and seem to have matured independently from each other [158]. This has been interpreted as a failure of IgM switching to IgG despite aberrant AID activity. However, only a partial overlapping of IgG and IgM populations has been demonstrated in various studies but it now needs to be replicated.

Naïve B‐cells are probably not randomly recruited in the inflammatory CNS since the VH family is biased [159]. Although the fate of naïve B‐cells emigrating to the CNS is still not clear, local maturation in the TLO structures and/or emigration to CLN are both probable (see below).

Most of the Ig peptides recovered from OCB by mass spectrometry match Ig‐secreting CSF B‐cells [14]. Moreover, clusters of related B‐cells present in the CSF are also sometimes recovered from blood, the largest bi‐compartmental proportion being observed in association with a recent relapse [14]. This observation may explain the existence of OCB “mirror” patterns 3 and 4 with overlapping B‐cell lineage on both sides of the BBB.

8.1. Local synthesis also occurs in the eye

Inflammatory lesions are observed in the eye in association with MS and perivenous lymphocyte cuffings (i.e., periphlebitis) are commonly observed, reminiscent of lymphoid aggregates [165]. Therefore, besides the classical intrathecal Ig synthesis, local intraocular synthesis may also affect the eyes and glands. The MRZ reaction occurs with single specificity in up to 76% of cases and with ≥2 specificities in 82% [166, 167], but the ratio of Fs between Fuchs heterochromic uveitis syndrome (FHUS) and MS is about 40‐fold, which is in the same range of ITS ratios as observed between MS and viral encephalitis. OCBs are always found in the CSF and aqueous humor of MS patients. Importantly, both OCB and MRZ patterns mismatch in most cases.

Advertisement

9. Meningeal tertiary lymphoid organs may play a role in B‐cell traffic and maturation

Meningeal TLOs are ectopic follicles with germinal centers aggregating a reticulum of CD35+ and CXCL13+ stromal/dendritic follicular cells, proliferating CD20+/Ki67+ B‐cells, Ig+ plasma/plasmablast cells, and CD138+ plasma cells [168]. The mantle zone is often lacking, whereas CD4+ and CD8+ T‐cells infiltrate follicles [169]. CXCL13 immunoreactivity is confined to dendritiform cells inside intra‐meningeal B‐cell follicles [163]. The abundance of plasma cells is variable and proliferating cells in B‐cell follicles, mostly CD20+ cells, are observed at rates varying from 0 to 43% [163]. CCL21 and adhesion molecule peripheral node addressin (PNAd), which selectively binds to naïve T and B lymphocytes and allows their homing to secondary lymphoid organs, are absent [163], suggesting that the homing is dependent on various markers specific to brain tissues (see reference [170]). TLO demonstration is associated with higher CSF cytokines (TNFα and IFNγ) [171] but further studies are needed. The functions of TLO have been well described in various models of peripheral inflammation. TLOs are able to mount a T‐cell memory response in connection with the other lymphoid compartments [172]. In these models, the amount of Ig synthesis occurring in TLO is completely uncoupled from the blood response, meaning that local Ig‐secreting cells are highly specific, although a nonspecific response is associated [173, 174]. In fact, TLOs commonly occur during EAE; AID is locally expressed in association with SHM and B‐cell switching occurs [175177]. The incidence of TLO in response to age insults is genetically determined in mice by two genes [178], but data are lacking in human and in EAE models.

Figure 5.

Schematic drawing of tertiary lymphoid organs (TLOs). A few TLOs are disseminated on the leptomeninges. At pathologic level, leptomeningeal TLOs are surrounded by subpial lesions (cell and myelin loss). The toxic mechanism is less certain and might involve TNFα secretion. At the immunological level, TLOs recruit naïve T‐ and B‐cells that undergo antigen presentation, AID expression, affinity maturation, and proliferation of B‐cells. Local Ig synthesis depends on plasmablasts/plasma cells and remains the only easily accessible bedside parameter indirectly informing about the presence of TLO (adapted from reference [179]).

In MS, the presence of TLO is associated with a more intense subpial demyelination (cortical lesions type III) and cell loss [169]. A strong topographical relationship is observed between TLO and subpial inflammation and an incidental observation showed that acute subpial lesions are associated with active local neurodegeneration (Figure 5) [180]. Owing to their small size and low number (about six follicles in positive blocks [168]), TLOs are probably largely underestimated by the sampling process of pathological examinations. Nevertheless, it was recently demonstrated that late post‐contrast FLAIR sequences on 3T‐MRI sequences might sample few leptomeningeal lesions (one lesion in 65% of patients, ≤6 in all of them) in a third of progressive patients [181], and this prevalence could be higher with 7T‐MRI. Pathological examination of one of them confirmed the congruence of the MRI lesion with TLO. Given the massive underestimation of TLO lesions, the practical consequences of this technique are currently being examined for their predictive clinical value. The longitudinal evolution of these structures and the effect of immunosuppressive therapies on them are unknown. Up to now, TLO could only be examined in the removed CNS structures including brain and leptomeninges but excluding skull and dura mater. Therefore, the recently discovered dura mater lymphatic network, which drains CSF to CLN, has never been examined in the context of MS. Since lymphatics are closely associated with TLO in all other tissues, confirmation of these structures might open up unexpected avenues in studies of inflammatory cell studies trafficking and maturation [182].

Advertisement

10. Epitope spreading

Epitope spreading in EAE and MS is mainly documented for T‐cells where it occurs in association with TLO formation [176]. The contribution of B‐cells to epitope spreading, especially for their intrathecal counterpart, is less well known. The intimate mechanisms driving epitope spreading are speculative and it remains unclear to what extent SHM contributes to this process.

It was observed some years ago that the ITS of IgG becomes enriched during the early phase after MS onset [27, 42, 6870, 74]. The frequency of the MRZ pattern increases, OCB‐negative patients become positive, and more OCBs occur in the few patients initially displaying a low number of OCB. Assessment of IgG synthesis with multiplex antigen arrays now allows epitope spreading against CNS antigens to be monitored over time. Serum studies have shown that both intramolecular and intermolecular spreading occurs early after the very first demyelinating index event in children but only in patients demonstrating a further clinical MS conversion [42]. By contrast, pediatric patients remaining monophasic fail to diversify their antigenic response. A failure to regulate the aberrant autoimmune response is tentatively thought to explain this observation. Studies on early harvested CSF would give rewarding clues about CNS B‐cell epitope spreading. A longitudinal description of the network of interactions in B‐ and T‐cell epitopes (functional immunomics) at both serum and CSF level, and their interaction with HLA polymorphisms, might shed light on the pathophysiology of MS [183].

11. The problem of Ig directed against nonspecific targets

11.1. The “MRZ pattern”

ITS may occur in supposedly healthy patients or in those suffering from nonimmune CNS disorders, but in such cases, synthesis is always directed against a former healed CNS infection such as Lyme disease, VZV, or HSV [22].

The long‐known “MRZ pattern” is an ITS of Ig directed against measles, rubella, and varicella zoster virus, the herpes virus sometimes being included (“MRZH”). Although the IgG fraction that belongs to this specific response in the CSF is estimated to represent less than 2% of the total amount of CSF IgG [78, 184, 185] and only a minor fraction of total OCB [77, 186, 187], the reaction is considered to be highly specific of MS. None of the MS patients in whom the MRZ pattern has been reported suffered from a history of clinical encephalitis involving these viruses. The proportion of MS patients with an elevated antibody index against ≥1 of these viruses increases over time and MS evolution [78]. This MRZ reaction is sometimes present in OCB‐negative MS patients and in CIS patients who will convert and it becomes more pronounced over time, as demonstrated by follow‐up LP [39, 188, 189].

ITS is polyspecific in a quarter of MS patients (against 2, 3, or 4 AI in 17, 4, and 2%), but remains monospecific (one elevated AI against M, R, Z, or H) in 22% [39]. The number of elevated AI strongly correlates with both age at spinal tap and disease duration and slightly changes over time [39, 190]. Moreover, there is a trend to a higher proportion of elevated AI in SP‐MS than in RR‐MS patients [191]. Immunosuppressive treatments including natalizumab are ineffective to prevent the persistence of the MRZ pattern [38, 103, 189]. Unlike in controls, no decline in AI levels occurs with age in MS patients considering either serum or CSF titers, and there is even a slight increase [192, 193].

11.2. Broadening the “MRZ pattern” to all encountered infectious agents?

A more comprehensive analysis of ITS with a larger range of infectious agents demonstrated that the MRZ pattern is an interesting concept that could be broadened. In fact, ITS against many other infectious agents has been confirmed in MS: rotavirus, HHV6 (20–30%), mumps, influenza, parainfluenza, adenovirus, respiratory syncytial virus, distemper virus, Coxsackie virus B4, vaccinia, JC virus (3%), CMV, poliovirus, toxoplasmosis (10%), Borrelia burgdorferi (26%), and mycoplasma (complete review in reference [194]). A previous silent CNS infection can be ruled out since the seroprevalence for each infection was the same in both MS and controls, whereas the latter had no specific reaction. Moreover, an ITS against tetanus and diphtheria toxoids was also demonstrated in vaccinated patients whose CNS was never exposed to native toxins [195, 196]. In a study of ITS against 17 infectious agents, 57% of MS patients had ITS against ≥5 infectious agents, with an increasing number over time [197], and virtually all patients demonstrated a reaction against at least one of nine viruses [198].

The pattern of reaction mirrors the individual's history of previous infections and the immunization level of the population: reaction against rubella is more observed in MS from Germany than in Cuban patients (lower incidence and immunization campaign in Cuban females) [199]. The rate of intrathecal reaction against a given infectious agent correlates with the rate of seroprevalence of this agent in the population.

Some major consequences can be drawn from these findings as follows:

  1. ITS against an infectious agent in MS considered in isolation should not be considered as a clue for a chronic infection in MS, since the coexistence of reaction against many infectious agents is the rule in MS.

  2. The concept of “MRZ pattern” should be extended to a “polyspecific infectious pattern.”

  3. The hypothesis of the antigenic mimicry of a single infectious agent should always be considered in the light of a broad reaction against various infectious agents, whose presence depends mostly on epidemiological variations.

Polyspecific ITS against all infectious agents is just a component of polyspecific ITS, which is thought to be characteristic of MS, and could throw light on the pathophysiology of MS.

11.3. Nonspecific synthesis against infectious agents is lower in MS than in CNS infections

The fraction of a specific antibody response against an infectious agent within the complete intrathecal IgG response is called the specific fraction (Fs ) [200]. For example, an Fs value for measles of 2% means that 2% of the total intrathecal IgG response is directed against measles. Neuroinfections are thought to be associated with very high Fs against viruses: Fs are 8–45% for viral encephalitis (HSV, measles, VZV, subacute sclerosing panencephalitis). In other words, about 55–92% of the intrathecal Ig in infectious pathologies are directed against non‐causative antigens [184, 200, 201], that is, a nonspecific ITS is very common even in neuroinfections. In MS, each specificity in the MRZH reaction typically retains a very low median Fs of 0.2–1.3% (ranging from 0.03 to 5.3) [200, 202, 203] and comparable results are obtained for Fs anti‐EBV [203]. These specific Fs results are about 40‐fold lower in MS patients than those found in neuroinfections without overlap.

The amount of intrathecally secreted specific antibodies should be proportional to the number of intrathecal plasma cells. If one considers that circulating plasma cells are nonspecifically and randomly selected from blood to home to CNS TLO, the relative proportion of each specific IgG synthesis in CSF should grossly parallel to their proportion in blood [197]. The ranking of specific antibody concentrations in blood and CSF differs in 67% of MS patients, confirming that CSF IgG secretion does not simply mirror blood secretion. ITS of the specific antibodies occurs independently from each other [200]. We propose two non‐mutually exclusive explanations. A first hypothesis involves the differential intrathecal proliferation of specific B‐cells after being recruited from blood but before being committed to terminal differentiation into plasma cells, owing to a favorable intrathecal lymphoid environment. A second hypothesis posits a nonrandom brain homing of circulating plasmablasts. It seems unlikely that plasmablast homing is driven by IgG specificity. Rather, the critical intensity of plasmablast/plasma cell recruitment to the brain owing to the intensity of the peripheral immune response to infection could be involved.

11.4. The paradox of low intrathecal anti‐EBV reaction

Seroprevalence for EBV is virtually complete in MS patients, so the probability of ITS should be at least equal to or higher than other nonspecific viruses. However, anti‐EBV intrathecal values have shown that an unexpectedly small proportion of patients have ITS (in the range of <20%) [203211]. Comparing CSF OCB against HHV6 and EBV, OCB occurred twice more frequently against HHV6 than against EBV [211]. Moreover, AI against EBV is sometimes twofold lower than AI against each MRZ component [207]. A difference in seroprevalence can be ruled out. Measles and rubella seroprevalence resulting from either natural infection or vaccination exceed 90% in most populations and vaccination campaigns started decades before [39]. Furthermore, varicella seroprevalence exceeds 90% in all European populations and EBV seroprevalence is virtually complete in MS patients.

The lower‐than‐expected intrathecal response against EBV is not consistent with the strong correlation linking high serum anti‐EBV levels and MS activity. In the light of this finding, such an extreme discrepancy can be interpreted as a strong clue for EBV infection preceding MS clinical onset [203]. Possible pathophysiological explanations have been developed elsewhere in the light of TLO [194]. The peculiar relationship between EBV and MS pathology is reinforced by the demonstration of a high intrathecal EBV‐specific CD8+ cytotoxic activity only early in MS patients, without recruitment of CD8+ cells against different targets (CMV‐specific CD8+ cells) [212] and the clearance of ITS in a case receiving autologous CD8+ T‐cells activated against EBV [109].

11.5. Nonspecific synthesis may be a simple property of meningeal tertiary lymphoid organs

Chronic nonspecific synthesis is constant in MS, but may not strictly indicate a bystanding reaction since they are almost absent in other mostly acute CNS inflammations [194]. These unspecific reactions rather indicate a dedicated specific function suggestive of the persistence of TLO in CNS. During immune activation in the periphery, naïve B‐cells undergo hypermutation of the Ig genes driven by germinal centers in the spleen and ganglia, and surviving cells are committed to plasmablasts released in the blood for a few days on their way to survival niches, where they differentiate into long‐lived plasma cells. Most of the niches are situated in the bone marrow but they are also to be found in secondary and tertiary lymphoid organs, where they display the ability to retain the newly formed plasma cells. Cultures of synovial fragments (containing TLO) from rheumatoid arthritis retain the ability to secrete non‐disease‐specific antibodies, such as anti‐tetanus toxin IgG [213]. Three weeks after, lupus‐prone mice were vaccinated against OVA, and the same number of antibody‐secreting cells against OVA was recovered in inflamed kidneys as in the bone marrow [214]. Therefore, nonspecific homing of plasma cells may be a common feature of inflammatory tissues and lead to local nonspecific synthesis.

The prolonged survival of retained plasma cells necessitates an anti‐apoptotic environment provided by cell niches. For example, CXCL12, which is a major determinant of PC retention in niches, is elevated in MS CSF, expressed by astrocytes and in the vicinity of lymphoid infiltrates [215]. Therefore, we previously suggested that nonspecific Ig synthesis in MS might simply be a common function of TLO harvesting Ig‐secreting cells, as is observed in most inflammatory disorders [194, 213]. An experimental demonstration was given by a series of MS patients who developed CSF IgG against TT after tetanus toxoid vaccination [196]. According to this assumption, the recruitment of intrathecal antibody‐secreting cells should be progressive and the complete MRZ pattern might take years or even decades to achieve. This was observed in clinical studies where the mean age at spinal tap and disease duration correlated with the number of elevated antibody indices [39]. Half of the patients having fewer than 5 years of disease duration had no elevated AI (MRZ), whereas 70% of those with more than 10 years duration had AI ≥2 [39]. A similar trend was observed in a study based on 17 antigens [197].

12. Deciphering a potentially disease‐related IgG ITS

12.1. MS‐specific targets? A puzzling problem

From an historical point of view, the quest for an Ig target in MS has followed the methodology successfully used to solve the paradigmatic problem of subacute sclerosing panencephalitis (SSPE), where it was demonstrated that the majority of intrathecal IgG was virus‐specific. Unfortunately, MS CSF demonstrated weak and inconsistent interaction with CNS antigens in numerous earlier studies. It was thereafter proposed that IgG production in MS may be due to nonsense antibodies [71]. These over‐hasty conclusions should now be mitigated.

CSF Igs constitute a private repertoire that may harbor toxic properties [216]. Functional electrophysiological modifications of neurotransmission after CSF application were observed long ago. Patterns of demyelinating lesions containing Ig and complement deposition in and around macrophages are typical of pattern 2 observed in MS [148, 217, 218]. The application of CSF to cultured cells (rat cerebellar granule neurons) significantly labels the axonal surface with IgM dots [118] and purified antibodies against myelin/oligodendrocyte glycoprotein (MOG) from MS serum bind to intact myelin in rat [219]. Various recombinant antibodies (rAb) synthesized from CD138+ CSF B‐cells of MS patients stain most of the glial components [220, 221] and many specific antibodies have been shown to react against cultured oligodendrocytes or human CNS tissue [222]. In animal models, anti‐MOG antibodies purified from human MS serum strongly enhance lesions without increasing inflammation [219, 223]. In a model of focal implantation of hybridoma cells secreting Ig against myelin in rat spinal cord, demyelination occurred around the implantation site [224]. Moreover, the implantation of anti‐GalC hybridoma cells may inhibit myelination in a complement‐dependent pathway both in vivo and in vitro [225, 226]. These latter models elegantly combine focal IgG synthesis with focal anatomical lesions and are reminiscent of the infiltration by plasma cells inside non‐remyelinated plaques.

In conclusion, the hypothesis that CSF Ig could play an active role in brain dysfunction, both at functional and at anatomical levels, is largely justified.

12.2. Does the microheterogeneity of ITS translate into disease heterogeneity?

Although the bulk of ITS targets unknown antigens, some of the antibody clones are thought to play an active biological role that could be either deleterious or beneficial. For example, occasional cases of MS may display anti‐N‐methyl‐D‐aspartate receptor (NMDAR) (<1%) or anti‐MOG (12%) antibodies, which are associated with cognitive and demyelinating features [227]. However, until now and unlike the anti‐aquaporin4 (AQP4) observed in neuromyelitis optica, none of the hundred or so identified antibodies has definitely been associated with the clinical (i.e., epilepsy, amnesia) or pathological distinctive features of MS (i.e., ability to remyelinate). The pathogenicity of these antibodies may be characterized in vitro by the extent of the demyelination or axonal loss obtained in myelinated cultures [216], by specific cell losses such as oligodendrocyte progenitors [228] or by functional alteration of synaptic transmission [89, 228]. For example, IgGs directed against MOG or sulfatide produce a complete myelin loss whereas the axons are preserved. On the other hand, IgG against neurofascine produces both myelin loss and loss of 15–40% of axons [216]. Therefore, at the global level, subsets of patients harbor variable toxicity against myelin or axons owing to one or more different antibody specificities against the targets. In the seminal work of Elliott et al. [216], a third of patients’ sera were able to destroy myelin and only 6% targeted axons, but this was probably massively underestimated owing to the limited number of targets examined, fewer than 10. Heterogeneity is also present at the clonal level since different IgG clones targeting the same antigens may display different pathogenicity ranging from null to high. This clonal microheterogeneity is driven by the ability of each clone to fix complement, and, for example, among the many MOG‐specific clones generated in EAE, only some of them also recognize the myelin sheath, and their pathogenicity is driven by complement fixation [187]. On the other end of the Ig spectrum, anti‐MBP IgMs are associated with a more benign course. Interestingly, these Ig target a shared epitope antigen between MBP and the extracellular loop of CD64, which is one of the IgG Fc receptors responsible for the immunologic properties of macrophages [229].

Microarrays using a small subset of autoantigens discriminated immune signatures in the sera of RR, SP, and PP MS patients [84]. Interestingly, immune signatures based on 13 IgG and one IgM also discriminated pathologic patterns I and II. These results assign a main role to Ig patterns in the pathophysiology of axonal loss, and replication in the CSF may be even more informative since paired serum and CSF samples display different patterns [93]. The large‐scale collection of targeted antigens would help to define the functional state of the intrathecal immune system. Moreover, the availability of techniques able to profile ITS IgG diversity and pathogenicity might help to decipher the crucial problem of predicting the severity of MS.

12.3. Putative target antigens—what do OCBs target inside the brain?

The targets are often considered to be largely unknown [230] and studies have yielded contradictory results. CSF antibodies were often considered to be nonspecific owing to their lack of specificity against the three major myelin proteins, but this does not preclude any other specificity and studies devoid of a priori may reveal unknown specificities against other membrane proteins, lipids, or glycolipids. The failure to find a major antigen for intra‐BBB‐synthesized Ig may not relate to a nonsense antibody production but instead may reflect the molecular complexity of the CNS and the presumed antigenic target [71]. By analogy with infectious diseases where the antigenic response is driven by pathogen antigenicity, most strategies have concentrated upon the identification of relevant antigenic targets. Paradoxically, from these efforts emerged the unexpected problem of the immense multiplicity and variability of targets.

12.3.1. A priori methods

In the last 30 years, the paradigmatic representation came by analogy with EAE models triggered by a single antigen vaccination, in line with Koch's postulates. Therefore, it was initially thought that ITS was directed against a (single) putative antigen. Unfortunately, intensive investigations of a wide range of potential targets were based on this misleading conception and failed to demonstrate any definitive association apart from more than 100 protein targets [231233] and roughly 300 epitopes (6% being non‐peptidic) [234]. Unlike in many other autoimmune disorders (e.g., myasthenia gravis), no study has succeeded in demonstrating any single antibody specificity for MS but few candidate epitopes (i.e., GAGA4) are still awaiting confirmation of their specificity by replication studies and the availability of commercial kits. Similar results have been obtained in SLE where large‐scale clusters of antibodies capture significant information regarding diagnosis and prognosis, whereas limited information is obtained from routinely measured antibodies [235].

12.3.2. Non‐a priori methods

12.3.2.1. Phage display

The phage display random peptide library was used to try to characterize unknown epitopes. Phagotopes identified from a CSF test pool of 10 MS patients were not recognized by the CSF of 55 other MS patients [76], which was interpreted as the demonstration that isolated phagotopes are patient‐ but not disease‐specific epitopes [76]. In a study using a similar design, 6 clones were selected from a CSF of 4 CIS, but panning to 187 MS patients found 0–22% positivity depending on the clone and MS type [236]. In the seminal work by Yu et al. [72], one dominant peptide was isolated from each of five MS CSF. Each peptide was patient‐specific, and in a given CSF, multiple OCBs (but not all) reacted against the peptide under study, confirming a restricted clonal IgG expression in CSF [72]. The specificity of CSF Ig against peptides was 10‐fold higher than from serum, suggesting a local affinity maturation of antibodies.

12.3.2.2. Generation of recombinant antibodies (rAbs) from clonally expanded plasma cells from CSF

The major advantages of this technique are as follows: (a) absence of a priori knowledge of the putative targets, (b) revelation of the most representative targeted peptides, and (c) precise identification of the candidate protein. Its main drawbacks are that it samples very few clones from each CSF. The generated rAbs clones bound to neuronal or astrocyte targets but myelin array failed to identify any precise target in most of them [237]. Interestingly, the aligned protein candidates identified among the multiple CSF analysis from the different groups did not overlap, thus suggesting a broad range of specificities [72, 238].

12.3.2.3. CSF analysis with protein microarrays

Basically, protein microarray chips are high‐throughput parallelized enzyme‐linked immunosorbent assay (ELISA) enabling extensive analysis of the breadth of antibody response. Sera from 17 RR‐MS patients were compared to those from 15 controls on a protein microarray printed with more than 3101 candidate antigens that represented multiple antigen families [231]. Although numerous candidate proteins were identified, no clue was obtained about individual pattern variability. A larger microarray displaying 37,000 tagged proteins was created in which patterns of immunoreactivity differed between individual patients [232]. The main drawback of this screening method is the biased antibody profiling approach, since the technique does not account for posttranslational modifications of proteins and non‐proteic targets.

12.4. Screening new targets associated with chronic CNS inflammation

Comparison of CSF Ig recognition patterns against normal or MS brain tissue lysates revealed a profile of 16 bands highly predictive of MS, suggesting that new antigens are specifically expressed in inflamed brain [239]. Chronic inflammation was associated with the aberrant expression of carbohydrate markers, which were associated with a high level of specific antibodies in CSF [240].

Development of microarrays targeting myelin proteome or brain lipids allowed the confirmation of a diverse polyclonal reaction against these non‐protidic targets both in humans and in EAE [84, 113, 241, 242]. Unconventional lipid targeting may be common since the proportion of CSF B‐cells targeting lipids was as high as 27% in a study based on rAb [243] and injection of anti‐sulfatid monoclonal antibody causes more severe EAE [241]. Unfortunately, only a limited number of potential targets were printed on these pioneering microarrays and only a minute number of rAb clones were obtained from a single patient.

Sugar moieties are an integral part of normal glycoproteins and glycolipids. Glycosylation occurs in the intracellular compartment where the first added sugar moieties are further masked in the mature‐released glycoprotein. Such “cryptic” carbohydrate structures are potentially immunogenic as demonstrated in human immunodeficiency virus (HIV)‐1 and cancer models [240]. Finally, an antigen microarray platform was constructed to display a wide range of autoantigens (proteins of the myelin sheath, liposomes of varying lipid composition, and cryptic glycan epitopes) and confirmed that CSF antibodies directed against various non‐proteinic structures are common in EAE and MS [240]. Although simultaneously targeting a limited number of structures, these pioneering studies opened perspectives for subsequent work based on larger microarrays [244, 245], thereby acknowledging the necessity to include various natural targets such as peptides, lipids, and glycans of both human and foreign origin and their posttranslational modifications. Interestingly, microbe‐derived polysaccharides are targeted by the host immune response and may share cross‐reactivity with normal human glycan structures [245]. In‐depth screening of foreign antigens including viral particles might be fruitful for deciphering a potential complex network of cross‐reactivity (Figure 6).

Figure 6.

Overview of antigen microarray principles. Interesting antigens are selected from libraries of autoantigen peptides or foreign peptides (i.e., EBV proteins), but post‐translationaly modified proteins, lipids, or glycans are suitable. Dense arrays of antigens are spotted on prepared glass slides. Patient samples (serum, CSF) are dropped on the chip, relevant antibodies hybridize to target antigens, and fluorescent‐labeled antibodies are applied for detection. The specific antibody signature results in a pattern informative of disease (MS vs other) and stages (RR vs SP) and reveals the early dynamics of the antibody response [42, 93]. Future chips may include a large variety of antigens including atypical (i.e., lipids, glycans, oxidized) and foreign antigens (i.e., viral proteins).

12.5. Posttranslational modifications

About 200 posttranslational modifications for proteins and sharing common properties have been described to date: PTMs target protein hotspots, mostly exposed on the protein surface, they may occur in physiologic life or in reaction to pathology, and protein modifications are irreversible. Although PTM‐targeting antibodies occurring in vivo have been rarely described until now in autoimmune disorders, they are being increasingly recognized and all of them might exist since most PTMs are targeted by commercially available monoclonal antibodies raised for research purposes. “Normal” antigens may be masked by PTM and may generate new self‐antigens. A growing body of evidence suggests that PTM may play a central role in some inflammatory pathologies such as rheumatoid arthritis [246], and in inflammatory bowel diseases, type I diabetes, and atherosclerosis. Tolerance to self‐antigens may be broken when self‐antigens undergo protein modifications, whereby they are recognized by antibodies and T‐cells as foreign antigens. This kind of molecular (self‐)mimicry might trigger inflammation. Therefore, PTM may also be key players for specific autoantibody recognition in MS. This major hypothesis could account for the failure to uncover specific targets of the intrathecally synthesized antibodies screened using native protein microarrays. Screening of libraries of synthetically posttranslational‐modified peptides mimicking antigenic epitopes may allow the characterization of new targets.

Citrullination is increased during EAE and MS and the targeted proteins are catabolized faster. Antibodies in rheumatoid arthritis mainly target citrullinated peptides. Methylation of arginine and lysine residues occurs in EAE brains and these PTMs are targeted in lupus erythematosus. Acetylation of intracellular proteins increases as MS evolves and acetylated MBP produces an immunodominant T‐cell epitope in mice that may trigger EAE [247].

Enzymatic glycosylation of protein is a common PTM and polymorphism of N‐glycan‐branching enzyme (MGAT5) is associated with the risk of MS and spontaneous EAE. The MOG fragment (aa30‐50) bears an N‐glycosylated asparagine at position 31 that is responsible for its antigenic properties and this PTM peptide is mimicked by the synthetic peptide CSF114(Glc) [248]. Another interesting candidate is the glucose‐based antigen GAGA4 [Glc(α1,4)Glc(α)] [249]. Both peptides are associated with specific serum antibodies in many MS patients.

Oxidization generates a large range of by‐products since reactive oxygen species (ROS) are able to oxidize nearly all the biological components according to their oxidation potential as follows:

  1. Oxidized proteins. Nitration of tyrosine residues, which normally occurs during brain aging and neurodegenerative processes, is specifically targeted by antibodies already identified during rheumatoid arthritis [246]. Malondialdehyde‐acetaldehyde adducts (MAA) are generated by the reaction of products of ROS‐mediated lipid peroxidation with peptidyl‐lysine. MAA adducts are immunogenic in the absence of adjuvant, abundant in synovial lesions of RA, and anti‐MAAs are elevated in rheumatoid arthritis and atherosclerosis patients [250]. Antibodies directed against oxidized proteins were demonstrated in the sera of EAE and SP‐MS patients [251].

  2. Oxidized sugars may form a stable advanced glycation end product that dramatically affects protein antigenicity since antibodies against the native form are usually non‐cross reactive with the PTM form [246].

  3. Oxidization of lipids may also create autoreactive antibodies. A high level of oxidation could be a distinctive feature of MS pathology in models of CNS inflammation and may be targeted by antibodies. Anti‐oxidized‐low‐density lipoprotein (LDL) antibodies have been described in the serum of MS patients. Oxidized phosphatidylcholine is more abundant in MS brain extracts and anti‐oxPCs are synthesized in the CSF of MS patients and EAE mice [252]. These results were confirmed in human MS and mice EAE with various lipid targets [241, 253]. The level of oxidative protein products in CSF does not influence the ITS level [254].

Considering the wide range of possibilities to modify self‐antigens by oxidation, this process is an attractive candidate to initiate an “oxidative PTM intolerance” [255].

12.6. Designing microarrays of PTM proteins for multiplex screening with antibodies from CSF of MS patients

Most of the PTM described above are induced by enzymatic catalysis or a chemical reaction occurring under stringent conditions, and their biologic synthesis is not facilitated by the large range of parameters involved. The question is whether it is possible to print large microarrays of antigens able to express all or most of the possible combinations of PTM. Technical considerations and successful experimentation on a small scale suggest that various PTMs may be successfully applied on immobilized proteins [255, 256] and that the development of commercial PTM protein microarrays holds much promise for the future.

13. Conclusion

A distinctive feature of MS is a long‐lasting intrathecal inflammatory response, unlike chronic CNS infections in which intrathecal inflammation abates after antibiotherapy. The next step will be to decipher the factors promoting the sustainment of the intrathecal inflammatory response and to identify specific antibody targets. Understanding the role of long‐lived plasma cells that produce OCB during the chronic CNS surge could open up new prospects for progressive MS therapies. As Tourtellotte stated long ago, one of the goals of an effective MS treatment could be the eradication of ITS. Therapies targeting CNS B‐cells to disrupt B‐cell traffic on both sides of the BBB are under development and eradication of resident CNS plasma cells remains an ultimate goal.

14. Abbreviations

  1. AI, activity index.

  2. AID, activation‐induced cytidine deaminase enzyme

  3. BBB, blood‐brain barrier

  4. CDR, complementary‐determining region

  5. CIS, clinically isolated syndrome

  6. CLN, cervical lymph node

  7. CMV, cytomegalovirus

  8. CNS, central nervous system

  9. CSF, cerebrospinal fluid

  10. EAE, experimental allergic encephalomyelitis

  11. EBV, Epstein‐Barr virus

  12. EDSS, expanded disability status scale

  13. FLC, free light chain

  14. HSV, herpes simplex virus

  15. IEF, isoelectric focusing

  16. Ig, immunoglobulin

  17. ITS, intrathecal (Ig) synthesis

  18. MS, multiple sclerosis

  19. OCB, oligoclonal bands

  20. PP, primary progressive MS

  21. PTM, posttranslational modification

  22. rAb, recombinant antibodies

  23. RR, relapsing‐remitting MS

  24. SHM, somatic hypermutation

  25. SP, secondary progressive MS

  26. TLO, tertiary lymphoid organs

  27. VZV, varicella zoster virus.

Acknowledgments

We are indebted to R. Cooke for copyediting and to Christel Baigts and Aurora Medevielle for bibliographical assistance.

References

  1. 1. 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(4): 465–76.
  2. 2. Anthony IC, Crawford DH, Bell JE. B lymphocytes in the normal brain: contrasts with HIV‐associated lymphoid infiltrates and lymphomas. Brain 2003; 126(Pt 5): 1058–67.
  3. 3. Doyle KP, Quach LN, Sole M, et al. B‐lymphocyte‐mediated delayed cognitive impairment following stroke. J Neurosci 2015; 35(5): 2133–45.
  4. 4. Felgenhauer K. Protein size and cerebrospinal fluid composition. Klin Wochenschr 1974; 52(24): 1158–64.
  5. 5. Reiber H. Dynamics of brain‐derived proteins in cerebrospinal fluid. Clin Chim Acta 2001; 310(2): 173–86.
  6. 6. Tourtellotte WW, Potvin AR, Fleming JO, et al. Multiple sclerosis: measurement and validation of central nervous system IgG synthesis rate. Neurology 1980; 30(3): 240–4.
  7. 7. Reiber H, Teut M, Pohl D, et al. Paediatric and adult multiple sclerosis: age‐related differences and time course of the neuroimmunological response in cerebrospinal fluid. Mult Scler 2009; 15(12): 1466–80.
  8. 8. Reiber H, Felgenhauer K. Protein transfer at the blood cerebrospinal fluid barrier and the quantitation of the humoral immune response within the central nervous system. Clin Chim Acta 1987; 163(3): 319–28.
  9. 9. Reiber H. Flow rate of cerebrospinal fluid (CSF)—a concept common to normal blood‐CSF barrier function and to dysfunction in neurological diseases. J Neurol Sci 1994; 122(2): 189–203.
  10. 10. Reiber H, Lange P. Quantification of virus‐specific antibodies in cerebrospinal fluid and serum: sensitive and specific detection of antibody synthesis in brain. Clin Chem 1991; 37(7): 1153–60.
  11. 11. Castillo‐Gomez E, Kastner A, Steiner J, et al. The brain as ‘immunoprecipitator’ of serum autoantibodies against NMDAR1. Ann Neurol 2016; 79(1):144–51
  12. 12. Rand KH, Houck H. Improved methods for the application of random peptide phage libraries to the study of the oligoclonal bands in cerebrospinal fluid of patients with multiple sclerosis. J Neurosci Methods 2000; 101(2): 131–9.
  13. 13. Stauch C, Reiber H, Rauchenzauner M, et al. Intrathecal IgM synthesis in pediatric MS is not a negative prognostic marker of disease progression: quantitative versus qualitative IgM analysis. Mult Scler 2011; 17(3): 327–34.
  14. 14. Bankoti J, Apeltsin L, Hauser SL, et al. In multiple sclerosis, oligoclonal bands connect to peripheral B‐cell responses. Ann Neurol 2014; 75(2): 266–76.
  15. 15. Obermeier B, Mentele R, Malotka J, et al. Matching of oligoclonal immunoglobulin transcriptomes and proteomes of cerebrospinal fluid in multiple sclerosis. Nat Med 2008; 14(6): 688–93.
  16. 16. Siden A, Kjellin KG. Isoelectric focusing of CSF and serum proteins in neurological disorders combined with benign and malignant proliferations of reticulocytes, lymphocytes and plasmocytes. J Neurol 1977; 216(4): 251–64.
  17. 17. Tourtellotte WW, Shapshak P, Staugaitis SM, et al. Hypothesis: multiple sclerosis cerebrospinal fluid IgG multiple bands are derived from one or a few IgG secretor cell clones. Prog Clin Biol Res 1984; 146(371-8).
  18. 18. Fasulo L, Fulgosi B, Colombatto S, et al. Uptake of polyamines by human lymphocytes. Ital J Biochem 1989; 38(4): 295A–6A.
  19. 19. Jefferis R. Glycosylation of recombinant antibody therapeutics. Biotechnol Prog 2005; 21(1): 11–6.
  20. 20. Halbgebauer S, Haussmann U, Klafki H, et al. Capillary isoelectric focusing immunoassay as a new nanoscale approach for the detection of oligoclonal bands. Electrophoresis 2015; 36(2): 355–62.
  21. 21. Dorries R, Watanabe R, Wege H, et al. Analysis of the intrathecal humoral immune response in Brown Norway (BN) rats, infected with the murine coronavirus JHM. J Neuroimmunol 1987; 14(3): 305–16.
  22. 22. Rostrom B, Link H, Laurenzi MA, et al. Viral antibody activity of oligoclonal and polyclonal immunoglobulins synthesized within the central nervous system in multiple sclerosis. Ann Neurol 1981; 9(6): 569–74.
  23. 23. Cruz M, Olsson T, Ernerudh J, et al. Immunoblot detection of oligoclonal anti‐myelin basic protein IgG antibodies in cerebrospinal fluid in multiple sclerosis. Neurology 1987; 37(9): 1515–9.
  24. 24. Bourahoui A, De Seze J, Guttierez R, et al. CSF isoelectrofocusing in a large cohort of MS and other neurological diseases. Eur J Neurol 2004; 11(8): 525–9.
  25. 25. Davies G, Keir G, Thompson E, et al. The clinical significance of an intrathecal monoclonal immunoglobulin band: a follow‐up study. Neurology 2003; 60:1163–6.
  26. 26. Freedman MS, Thompson EJ, Deisenhammer F, et al. Recommended standard of cerebrospinal fluid analysis in the diagnosis of multiple sclerosis: a consensus statement. Arch Neurol 2005; 62(6): 865–70.
  27. 27. Villar LM, Masterman T, Casanova B, et al. CSF oligoclonal band patterns reveal disease heterogeneity in multiple sclerosis. J Neuroimmunol 2009; 211(1–2): 101–4.
  28. 28. Andersson M, Alvarez‐Cermeno J, Bernardi G, et al. Cerebrospinal fluid in the diagnosis of multiple sclerosis: a consensus report. J Neurol Neurosurg Psychiatr 1994; 57(8): 897–902.
  29. 29. Jarius S, Eichhorn P, Wildemann B, et al. Usefulness of antibody index assessment in cerebrospinal fluid from patients negative for total‐IgG oligoclonal bands. Fluids Barriers CNS 2012; 9(1): 14.
  30. 30. Lolli F, Halawa I, Link H. Intrathecal synthesis of IgG, IgA, IgM and IgD in untreated multiple sclerosis and controls. Acta Neurol Scand 1989; 80(3): 238–47.
  31. 31. Sena A, Rosado P, Ferret‐Sena V, et al. Multiple sclerosis and intrathecal IgA synthesis. Acta Neurol Belg 1997; 97(1): 36–8.
  32. 32. Kaplan B, Aizenbud BM, Golderman S, et al. Free light chain monomers in the diagnosis of multiple sclerosis. J Neuroimmunol 2010; 229(1–2): 263–71.
  33. 33. Sindic CJ, Laterre EC. Oligoclonal free kappa and lambda bands in the cerebrospinal fluid of patients with multiple sclerosis and other neurological diseases. An immunoaffinity‐mediated capillary blot study. J Neuroimmunol 1991; 33(1): 63–72.
  34. 34. Goffette S, Schluep M, Henry H, et al. Detection of oligoclonal free kappa chains in the absence of oligoclonal IgG in the CSF of patients with suspected multiple sclerosis. J Neurol Neurosurg Psychiatr 2004; 75(2): 308–10.
  35. 35. Presslauer S, Milosavljevic D, Huebl W, et al. Validation of kappa free light chains as a diagnostic biomarker in multiple sclerosis and clinically isolated syndrome: a multicenter study. Mult Scler 2015;22(4):502-10.
  36. 36. Stich O, Kluge J, Speck J, et al. Oligoclonal restriction of antiviral immunoreaction in oligoclonal band‐negative MS patients. Acta Neurol Scand. 2015; 131(6):381-8.
  37. 37. Bednárová J, Stourac P, Adam P. Relevance of immunological variables in neuroborreliosis and multiple sclerosis. Acta Neurol Scand 2005; 112(2): 97–102.
  38. 38. Jarius S, Franciotta D, Bergamaschi R, et al. Polyspecific, antiviral immune response distinguishes multiple sclerosis and neuromyelitis optica. J Neurol Neurosurg Psychiatr 2008; 79(10): 1134–6.
  39. 39. Brecht I, Weissbrich B, Braun J, et al. Intrathecal, polyspecific antiviral immune response in oligoclonal band negative multiple sclerosis. PLoS One 2012; 7(7): e40431.
  40. 40. Frederiksen JL, Sindic CJ. Intrathecal synthesis of virus‐specific oligoclonal IgG, and of free kappa and free lambda oligoclonal bands in acute monosymptomatic optic neuritis. Comparison with brain MRI. Mult Scler 1998; 4(1): 22–6.
  41. 41. Stich O, Kluge J, Speck J, et al. Detection of virus‐specific (measles, rubella, zoster) oligoclonal IgG‐bands in CSF from multiple sclerosis patients without oligoclonal bands of total IgG. Mult Scler 2009; 15(S86).
  42. 42. Quintana FJ, Patel B, Yeste A, et al. Epitope spreading as an early pathogenic event in pediatric multiple sclerosis. Neurology 2014; 83(24): 2219–26.
  43. 43. Cabre P, Heinzlef O, Merle H, et al. MS and neuromyelitis optica in Martinique (French West Indies). Neurology 2001; 56(4): 507–14.
  44. 44. Yoshimura S, Isobe N, Matsushita T, et al. Genetic and infectious profiles influence cerebrospinal fluid IgG abnormality in Japanese multiple sclerosis patients. PLoS One 2014; 9(4): e95367.
  45. 45. Cross AH, Stark JL, Lauber J, et al. Rituximab reduces B cells and T cells in cerebrospinal fluid of multiple sclerosis patients. J Neuroimmunol 2006; 180(1–2): 63–70.
  46. 46. Izquierdo G, Angulo S, Garcia‐Moreno JM, et al. Intrathecal IgG synthesis: marker of progression in multiple sclerosis patients. Acta Neurol Scand 2002; 105(3): 158–63.
  47. 47. Rudick RA, Medendorp SV, Namey M, et al. Multiple sclerosis progression in a natural history study: predictive value of cerebrospinal fluid free kappa light chains. Mult Scler 1995; 1(3): 150–5.
  48. 48. da Gama PD, Machado Ldos R, Livramento JA, et al. Oligoclonal bands in cerebrospinal fluid of black patients with multiple sclerosis. Biomed Res Int 2015; 2015(217961).
  49. 49. Rinker JR, 2nd, Trinkaus K, Naismith RT, et al. Higher IgG index found in African Americans versus Caucasians with multiple sclerosis. Neurology 2007; 69(1): 68–72.
  50. 50. Chang KH, Lyu RK, Chen CM, et al. Clinical characteristics of multiple sclerosis in Taiwan: a cross‐sectional study. Mult Scler 2006; 12(4): 501–6.
  51. 51. Kim SH, Huh SY, Kim W, et al. Clinical characteristics and outcome of multiple sclerosis in Korea: does multiple sclerosis in Korea really differ from that in the Caucasian populations? Mult Scler 2013; 19(11): 1493–8.
  52. 52. Li B, Dong H, Zhang J, et al. Cerebrospinal fluid IgG profiles and oligoclonal bands in Chinese patients with multiple sclerosis. Acta Neurol Scand 2007; 115(5): 319–24.
  53. 53. Niino M, Sato S, Fukazawa T, et al. Latitude and HLA‐DRB1 alleles independently affect the emergence of cerebrospinal fluid IgG abnormality in multiple sclerosis. Mult Scler 2015; 21(9): 1112–20.
  54. 54. Sellebjerg F, Jensen J, Madsen HO, et al. HLA DRB1*1501 and intrathecal inflammation in multiple sclerosis. Tissue Antigens 2000; 55(4): 312–8.
  55. 55. Gama PD, Machado Ldos R, Livramento JA, et al. Study of oligoclonal bands restricted to the cerebrospinal fluid in multiple sclerosis patients in the city of Sao Paulo. Arq Neuropsiquiatr 2009; 67(4): 1017–22.
  56. 56. Leone MA, Barizzone N, Esposito F, et al. Association of genetic markers with CSF oligoclonal bands in multiple sclerosis patients. PLoS One 2013; 8(6): e64408.
  57. 57. Mero IL, Gustavsen MW, Saether HS, et al. Oligoclonal band status in Scandinavian multiple sclerosis patients is associated with specific genetic risk alleles. PLoS One 2013; 8(3): e58352.
  58. 58. Pandey JP, Li Z. The forgotten tale of immunoglobulin allotypes in cancer risk and treatment. Exp Hematol Oncol 2013; 2(1): 6.
  59. 59. Raposo B, Dobritzsch D, Ge C, et al. Epitope‐specific antibody response is controlled by immunoglobulin V(H) polymorphisms. J Exp Med 2014; 211(3): 405–11.
  60. 60. Lefranc MP, Lefranc G. Human Gm, Km, and Am allotypes and their molecular characterization: a remarkable demonstration of polymorphism. Methods Mol Biol 2012; 882:635–80.
  61. 61. Raknes G, Fernandes Filho JA, Pandey JP, et al. IgG allotypes and subclasses in Norwegian patients with multiple sclerosis. J Neurol Sci 2000; 175(2): 111–5.
  62. 62. Sandberg‐Wollheim M, Baird LG, Schanfield MS, et al. Association of CSF IgG concentration and immunoglobulin allotype in multiple sclerosis and optic neuritis. Clin Immunol Immunopathol 1984; 31(2): 212–21.
  63. 63. Goris A, Pauwels I, Gustavsen MW, et al. Genetic variants are major determinants of CSF antibody levels in multiple sclerosis. Brain 2015; 138(Pt 3): 632–43.
  64. 64. Persson L, Longhi S, Enarsson J, et al. Elevated antibody reactivity to measles virus NCORE protein among patients with multiple sclerosis and their healthy siblings with intrathecal oligoclonal immunoglobulin G production. J Clin Virol 2014; 61(1):107–12.
  65. 65. Poser CM. Multiple sclerosis trait: the premorbid stage of multiple sclerosis. A hypothesis. Acta Neurol Scand 2004; 109(4): 239–43.
  66. 66. Duquette P, Charest L. Cerebrospinal fluid findings in healthy siblings of multiple sclerosis patients. Neurology 1986; 36(5): 727–9.
  67. 67. Haghighi S, Andersen O, Rosengren L, et al. Incidence of CSF abnormalities in siblings of multiple sclerosis patients and unrelated controls. J Neurol 2000; 247(8): 616–22.
  68. 68. Ghezzi A, Pozzilli C, Liguori M, et al. Prospective study of multiple sclerosis with early onset. Mult Scler 2002; 8(2): 115–8.
  69. 69. Pohl D, Rostasy K, Reiber H, et al. CSF characteristics in early‐onset multiple sclerosis. Neurology 2004; 63(10): 1966–7.
  70. 70. Zeman AZ, Kidd D, McLean BN, et al. A study of oligoclonal band negative multiple sclerosis. J Neurol Neurosurg Psychiatr 1996; 60(1): 27–30.
  71. 71. Walsh MJ, Tourtellotte WW. Temporal invariance and clonal uniformity of brain and cerebrospinal IgG, IgA, and IgM in multiple sclerosis. J Exp Med 1986; 163(1): 41–53.
  72. 72. Yu X, Burgoon M, Green M, et al. Intrathecally synthesized IgG in multiple sclerosis cerebrospinal fluid recognizes identical epitopes over time. J Neuroimmunol 2011; 240–241: 129–36.
  73. 73. Villar LM, Masjuan J, González‐Porqué P, et al. Intrathecal IgM synthesis is a prognostic factor in multiple sclerosis. Ann Neurol 2003; 53(2): 222–6.
  74. 74. Owens GP, Ritchie AM, Burgoon MP, et al. Single‐cell repertoire analysis demonstrates that clonal expansion is a prominent feature of the B cell response in multiple sclerosis cerebrospinal fluid. J Immunol 2003; 171(5): 2725–33.
  75. 75. Colombo M, Dono M, Gazzola P, et al. Maintenance of B lymphocyte‐related clones in the cerebrospinal fluid of multiple sclerosis patients. Eur J Immunol 2003; 33(12): 3433–8.
  76. 76. Cortese I, Capone S, Luchetti S, et al. CSF‐enriched antibodies do not share specificities among MS patients. Mult Scler 1998; 4(3): 118–23.
  77. 77. Derfuss T, Gurkov R, Then Bergh F, et al. Intrathecal antibody production against Chlamydia pneumoniae in multiple sclerosis is part of a polyspecific immune response. Brain 2001; 124(Pt 7): 1325–35.
  78. 78. Reiber H, Ungefehr S, Jacobi C. The intrathecal, polyspecific and oligoclonal immune response in multiple sclerosis. Mult Scler 1998; 4(3): 111–7.
  79. 79. > Kuenz B, Lutterotti A, Ehling R, et al. Cerebrospinal fluid B cells correlate with early brain inflammation in multiple sclerosis. PLoS ONE 2008; 3(7):e2559
  80. 80. Lamers KJ, de Reus HP, Jongen PJ. Myelin basic protein in CSF as indicator of disease activity in multiple sclerosis. Mult Scler 1998; 4(3): 124–6.
  81. 81. Lamers KJ, Uitdehaag BM, Hommes OR, et al. The short‐term effect of an immunosuppressive treatment on CSF myelin basic protein in chronic progressive multiple sclerosis. J Neurol Neurosurg Psychiatr 1988; 51(10): 1334–7.
  82. 82. Confavreux C, Chapuis‐Cellier C, Arnaud P, et al. Oligoclonal “fingerprint” of CSF IgG in multiple sclerosis patients is not modified following intrathecal administration of natural beta‐interferon. J Neurol Neurosurg Psychiatr 1986; 49(11): 1308–12.
  83. 83. Tourtellotte WW, Baumhefner RW, Syndulko K, et al. The long march of the cerebrospinal fluid profile indicative of clinical definite multiple sclerosis; and still marching. J Neuroimmunol 1988; 20(2–3): 217–27.
  84. 84. Quintana FJ, Farez MF, Viglietta V, et al. Antigen microarrays identify unique serum autoantibody signatures in clinical and pathologic subtypes of multiple sclerosis. Proc Natl Acad Sci U S A 2008; 105(48): 18889–94.
  85. 85. Meinl E, Krumbholz M, Derfuss T, et al. Compartmentalization of inflammation in the CNS: a major mechanism driving progressive multiple sclerosis. J Neurol Sci 2008; 274(1–2): 42–4.
  86. 86. Bonnan M. Intrathecal IgG synthesis: a resistant and valuable target for future multiple sclerosis treatments. Mult Scler Int 2014; 2015:296184.
  87. 87. Butler WT, Rossen RD. Effects of corticosteroids on immunity in man. I. Decreased serum IgG concentration caused by 3 or 5 days of high doses of methylprednisolone. J Clin Invest 1973; 52(10): 2629–40.
  88. 88. Baumhefner RW, Booe MM, Tourtellotte WW. Modulation of de novo CNS IgG synthesis with preservation of oligoclonal IgG in multiple sclerosis. Neurology 1979; 29(4):549.
  89. 89. Frequin ST, Barkhof F, Lamers KJ, et al. CSF myelin basic protein, IgG and IgM levels in 101 MS patients before and after treatment with high‐dose intravenous methylprednisolone. Acta Neurol Scand 1992; 86(3): 291–7.
  90. 90. Trotter JL, Garvey WF. Prolonged effects of large‐dose methylprednisolone infusion in multiple sclerosis. Neurology 1980; 30(7 Pt 1): 702–8.
  91. 91. Tourtellotte WW, Baumhefner RW, Potvin AR, et al. Multiple sclerosis de novo CNS IgG synthesis: effect of ACTH and corticosteroids. Neurology 1980; 30(11): 1155–62.
  92. 92. Miro J, Aguayo F, Garrido JC, et al. Intrathecal immunoglobulin synthesis in multiple sclerosis: effect of corticosteroids and azathioprine. Eur Neurol 1992; 32(6): 349–53.
  93. 93. Quintana F, Rahbari R, Magalhaes S, et al. Specific Serum Antibody Patterns Detected with Antigen Arrays Are Associated to the Development of MS in Pediatric Patients (S60.006). Neurology 2012; 78(Meeting Abstracts 1): S60.006–S60.
  94. 94. Piccio L, Naismith RT, Trinkaus K, et al. Changes in B‐ and T‐lymphocyte and chemokine levels with rituximab treatment in multiple sclerosis. Arch Neurol 2010; 67(6): 707–14.
  95. 95. Hawker K, O'Connor P, Freedman MS, et al. Rituximab in patients with primary progressive multiple sclerosis: results of a randomized double‐blind placebo‐controlled multicenter trial. Ann Neurol 2009; 66(4): 460–71.
  96. 96. Bonnan M, Ferrari S, Bertandeau E, et al. Intrathecal rituximab therapy in multiple sclerosis: review of evidence supporting the need for future trials. Curr Drugs Targets 2014; 15(13): 1205–14.
  97. 97. Rossi S, Motta C, Studer V, et al. Tumor necrosis factor is elevated in progressive multiple sclerosis and causes excitotoxic neurodegeneration. Mult Scler 2014; 20(3): 304–12.
  98. 98. Studer V, Rossi S, Motta C, et al. Peripheral B cell depletion and central proinflammatory cytokine reduction following repeated intrathecal administration of rituximab in progressive multiple sclerosis. J Neuroimmunol 2014;276(1-2):229-31.
  99. 99. Harrer A, Tumani H, Niendorf S, et al. Cerebrospinal fluid parameters of B cell‐related activity in patients with active disease during natalizumab therapy. Mult Scler 2013; 19(9): 1209–12.
  100. 100. Mancuso R, Franciotta D, Rovaris M, et al. Effects of natalizumab on oligoclonal bands in the cerebrospinal fluid of multiple sclerosis patients: a longitudinal study. Mult Scler 2014;20(14):1900-3.
  101. 101. von Glehn F, Farias AS, de Oliveira AC, et al. Disappearance of cerebrospinal fluid oligoclonal bands after natalizumab treatment of multiple sclerosis patients. Mult Scler 2012; 18(7): 1038–41.
  102. 102. Romme Christensen J, Ratzer R, Bornsen L, et al. Natalizumab in progressive MS: results of an open‐label, phase 2A, proof‐of‐concept trial. Neurology 2014; 82(17): 1499–507.
  103. 103. Warnke C, Stettner M, Lehmensiek V, et al. Natalizumab exerts a suppressive effect on surrogates of B cell function in blood and CSF. Mult Scler 2014;21(8):1036-44.
  104. 104. Withers DR, Fiorini C, Fischer RT, et al. T cell‐dependent survival of CD20+ and CD20‐ plasma cells in human secondary lymphoid tissue. Blood 2007; 109(11): 4856–64.
  105. 105. de Andres C, Teijeiro R, Alonso B, et al. Long‐term decrease in VLA‐4 expression and functional impairment of dendritic cells during natalizumab therapy in patients with multiple sclerosis. PLoS One 2012; 7(4): e34103.
  106. 106. Martin M, Cravens PD, Winger R, et al. Decrease in the numbers of dendritic cells and CD4+ T cells in cerebral perivascular spaces due to natalizumab. Arch Neurol 2008; 65(12): 1596–603.
  107. 107. Selter RC, Biberacher V, Grummel V, et al. Natalizumab treatment decreases serum IgM and IgG levels in multiple sclerosis patients. Mult Scler 2013; 19(11): 1454–61.
  108. 108. Stuve O, Cravens PD, Frohman EM, et al. Immunologic, clinical, and radiologic status 14 months after cessation of natalizumab therapy. Neurology 2009; 72(5): 396–401.
  109. 109. Pender MP, Csurhes PA, Smith C, et al. Epstein‐Barr virus‐specific adoptive immunotherapy for progressive multiple sclerosis. Mult Scler 2014;20(11):1541-4.
  110. 110. Di Pauli F, Gredler V, Kuenz B, et al. Features of intrathecal immunoglobulins in patients with multiple sclerosis. J Neurol Sci 2010; 288(1–2): 147–50.
  111. 111. Tintore M, Rovira A, Rio J, et al. Do oligoclonal bands add information to MRI in first attacks of multiple sclerosis? Neurology 2008; 70(13 Pt 2): 1079–83.
  112. 112. Dalla Costa G, Passerini G, Messina MJ, et al. Clinical significance of the number of oligoclonal bands in patients with clinically isolated syndromes. J Neuroimmunol. 2015; 289:62-7.
  113. 113. Villar LM, Sadaba MC, Roldan E, et al. Intrathecal synthesis of oligoclonal IgM against myelin lipids predicts an aggressive disease course in MS. J Clin Invest 2005; 115(1): 187–94.
  114. 114. Boscá I, Magraner MJ, Coret F, et al. The risk of relapse after a clinically isolated syndrome is related to the pattern of oligoclonal bands. J Neuroimmunol 2010; 226(1–2): 143–6.
  115. 115. Tumani H, Tourtellotte WW, Peter JB, et al. Acute optic neuritis: combined immunological markers and magnetic resonance imaging predict subsequent development of multiple sclerosis. The Optic Neuritis Study Group. J Neurol Sci 1998; 155(1): 44–9.
  116. 116. Rinker JR, 2nd, Trinkaus K, Cross AH. Elevated CSF free kappa light chains correlate with disability prognosis in multiple sclerosis. Neurology 2006; 67(7): 1288–90.
  117. 117. Villar LM, Masjuan J, Gonzalez‐Porque P, et al. Intrathecal IgM synthesis in neurologic diseases: relationship with disability in MS. Neurology 2002; 58(5): 824–6.
  118. 118. Beltrán E, Hernández A, Lafuente EM, et al. Neuronal antigens recognized by cerebrospinal fluid IgM in multiple sclerosis. J Neuroimmunol 2012; 247(1–2): 63–9.
  119. 119. Magraner MJ, Bosca I, Simó‐Castelló M, et al. Brain atrophy and lesion load are related to CSF lipid‐specific IgM oligoclonal bands in clinically isolated syndromes. Neuroradiology 2012; 54(1): 5–12.
  120. 120. Bosca I, Villar LM, Coret F, et al. Response to interferon in multiple sclerosis is related to lipid‐specific oligoclonal IgM bands. Mult Scler (Houndmills, Basingstoke, England) 2010; 16(7): 810–5.
  121. 121. Garcia‐Barragan N, Villar LM, Espino M, et al. Multiple sclerosis patients with anti‐lipid oligoclonal IgM show early favourable response to immunomodulatory treatment. Eur J Neurol 2009; 16(3): 380–5.
  122. 122. Mandrioli J, Sola P, Bedin R, et al. A multifactorial prognostic index in multiple sclerosis. Cerebrospinal fluid IgM oligoclonal bands and clinical features to predict the evolution of the disease. J Neurol 2008; 255(7): 1023–31.
  123. 123. Thangarajh M, Gomez‐Rial J, Hedstrom AK, et al. Lipid‐specific immunoglobulin M in CSF predicts adverse long‐term outcome in multiple sclerosis. Mult Scler 2008; 14(9): 1208–13.
  124. 124. Perini P, Ranzato F, Calabrese M, et al. Intrathecal IgM production at clinical onset correlates with a more severe disease course in multiple sclerosis. J Neurol Neurosurg Psychiatr 2006; 77(8): 953–5.
  125. 125. Schneider R, Euler B, Rauer S. Intrathecal IgM‐synthesis does not correlate with the risk of relapse in patients with a primary demyelinating event. Eur J Neurol 2007; 14(8): 907–11.
  126. 126. Imrell K, Landtblom A‐M, Hillert J, et al. Multiple sclerosis with and without CSF bands: clinically indistinguishable but immunogenetically distinct. Neurology 2006; 67(6): 1062–4.
  127. 127. Avasarala JR, Cross AH, Trotter JL. Oligoclonal band number as a marker for prognosis in multiple sclerosis. Arch Neurol 2001; 58(12): 2044–5.
  128. 128. Annunziata P, Giorgio A, De Santi L, et al. Absence of cerebrospinal fluid oligoclonal bands is associated with delayed disability progression in relapsing‐remitting MS patients treated with interferon‐beta. J Neurol Sci 2006; 244(1–2): 97–102.
  129. 129. Lechner‐Scott J, Spencer B, de Malmanche T, et al. The frequency of CSF oligoclonal banding in multiple sclerosis increases with latitude. Mult Scler 2012; 18(7): 974–82.
  130. 130. Rojas JI, Tizio S, Patrucco L, et al. Oligoclonal bands in multiple sclerosis patients: worse prognosis? Neurol Res 2012; 34(9): 889–92.
  131. 131. Siritho S, Freedman MS. The prognostic significance of cerebrospinal fluid in multiple sclerosis. J Neurol Sci 2009; 279(1–2): 21–5.
  132. 132. Joseph FG, Hirst CL, Pickersgill TP, et al. CSF oligoclonal band status informs prognosis in multiple sclerosis: a case control study of 100 patients. J Neurol Neurosurg Psychiatr 2009; 80(3): 292–6.
  133. 133. Cepok S, Rosche B, Grummel V, et al. Short‐lived plasma blasts are the main B cell effector subset during the course of multiple sclerosis. Brain 2005; 128(Pt 7): 1667–76.
  134. 134. Bonnan M, Barroso B, Demasles S, et al. Compartmentalized intrathecal immunoglobulin synthesis during HIV infection: a model of chronic CNS inflammation? J Neuroimmunol 2015; 285:41-52.
  135. 135. Bernheimer H, Lassmann H, Suchanek G. Dynamics of IgG+, IgA+, and IgM+ plasma cells in the central nervous system of guinea pigs with chronic relapsing experimental allergic encephalomyelitis. Neuropathol Appl Neurobiol 1988; 14(2): 157–67.
  136. 136. Dorries R, Schwender S, Imrich H, et al. Population dynamics of lymphocyte subsets in the central nervous system of rats with different susceptibility to coronavirus‐induced demyelinating encephalitis. Immunology 1991; 74(3): 539–45.
  137. 137. Rainey‐Barger EK, Rumble JM, Lalor SJ, et al. The lymphoid chemokine, CXCL13, is dispensable for the initial recruitment of B cells to the acutely inflamed central nervous system. Brain Behav Immun 2011; 25(5): 922–31.
  138. 138. Choi SR, Howell OW, Carassiti D, et al. Meningeal inflammation plays a role in the pathology of primary progressive multiple sclerosis. Brain 2012; 135(Pt 10): 2925–37.
  139. 139. Iivanainen M, Driscoll B, Richert J, et al. Oligoclonal IgG in the cerebrospinal fluid of guinea pigs with experimental allergic encephalomyelitis. Proc Soc Exp Biol Med 1982; 171(3): 272–5.
  140. 140. Mehta PD, Lassmann H, Wisniewski HM. Immunologic studies of chronic relapsing EAE in guinea pigs: similarities to multiple sclerosis. J Immunol 1981; 127(1): 334–8.
  141. 141. Pachner AR, Li L, Lagunoff D. Plasma cells in the central nervous system in the Theiler's virus model of multiple sclerosis. J Neuroimmunol 2011; 232(1–2): 35–40.
  142. 142. Walls AF, Suckling AJ, Rumsby MG. The origin and specificity of intrathecal IgG in chronic relapsing experimental allergic encephalomyelitis. Autoimmunity 1989; 2(2): 123–32.
  143. 143. Dorries R, Liebert UG, ter Meulen V. Comparative analysis of virus‐specific antibodies and immunoglobulins in serum and cerebrospinal fluid of subacute measles virus‐induced encephalomyelitis (SAME) in rats and subacute sclerosing panencephalitis (SSPE). J Neuroimmunol 1988; 19(4): 339–52.
  144. 144. Cserr HF, DePasquale M, Harling‐Berg CJ, et al. Afferent and efferent arms of the humoral immune response to CSF‐administered albumins in a rat model with normal blood‐brain barrier permeability. J Neuroimmunol 1992; 41(2): 195–202.
  145. 145. Furr M. Humoral immune responses in the horse after intrathecal challenge with ovalbumin. J Vet Intern Med 2007; 21(4): 806–11.
  146. 146. Gordon LB, Knopf PM, Cserr HF. Ovalbumin is more immunogenic when introduced into brain or cerebrospinal fluid than into extracerebral sites. J Neuroimmunol 1992; 40(1): 81–7.
  147. 147. Greve B, Magnusson CG, Melms A, et al. Immunoglobulin isotypes reveal a predominant role of type 1 immunity in multiple sclerosis. J Neuroimmunol 2001; 121(1–2): 120–5.
  148. 148. Zhang Y, Da R‐R, Hilgenberg LG, et al. Clonal expansion of IgA‐positive plasma cells and axon‐reactive antibodies in MS lesions. J Neuroimmunol 2005; 167(1–2): 120–30.
  149. 149. Stern JN, Yaari G, Vander Heiden JA, et al. B cells populating the multiple sclerosis brain mature in the draining cervical lymph nodes. Sci Transl Med 2014; 6(248): 248ra107.
  150. 150. Sindic CJ, Monteyne P, Bigaignon G, et al. Polyclonal and oligoclonal IgA synthesis in the cerebrospinal fluid of neurological patients: an immunoaffinity‐mediated capillary blot study. J Neuroimmunol 1994; 49(1–2): 109–14.
  151. 151. Nakano T, Matsui M, Inoue I, et al. Free immunoglobulin light chain: its biology and implications in diseases. Clin Chim Acta 2011; 412(11–12): 843–9.
  152. 152. Hassan‐Smith G, Durant L, Tsentemeidou A, et al. High sensitivity and specificity of elevated cerebrospinal fluid kappa free light chains in suspected multiple sclerosis. J Neuroimmunol 2014; 276(1–2): 175–9.
  153. 153. Duranti F, Pieri M, Centonze D, et al. Determination of kappaFLC and kappa Index in cerebrospinal fluid: a valid alternative to assess intrathecal immunoglobulin synthesis. J Neuroimmunol 2013; 263(1–2): 116–20.
  154. 154. Mehta PD. Quantitation of IgG subclasses in cerebrospinal fluid of patients with multiple sclerosis. Ann N Y Acad Sci 1988; 540:261–3.
  155. 155. Bennett JL, Haubold K, Ritchie AM, et al. CSF IgG heavy‐chain bias in patients at the time of a clinically isolated syndrome. J Neuroimmunol 2008; 199(1–2): 126–32.
  156. 156. Colombo M, Dono M, Gazzola P, et al. Accumulation of clonally related B lymphocytes in the cerebrospinal fluid of multiple sclerosis patients. J Immunol 2000; 164(5): 2782–9.
  157. 157. Owens GP, Winges KM, Ritchie AM, et al. VH4 gene segments dominate the intrathecal humoral immune response in multiple sclerosis. J Immunol 2007; 179(9): 6343–51.
  158. 158. Beltran E, Obermeier B, Moser M, et al. Intrathecal somatic hypermutation of IgM in multiple sclerosis and neuroinflammation. Brain 2014; 137(Pt 10): 2703–14.
  159. 159. Palanichamy A, Apeltsin L, Kuo TC, et al. Immunoglobulin class‐switched B cells form an active immune axis between CNS and periphery in multiple sclerosis. Sci Transl Med 2014; 6(248): 248ra106.
  160. 160. Cameron EM, Spencer S, Lazarini J, et al. Potential of a unique antibody gene signature to predict conversion to clinically definite multiple sclerosis. J Neuroimmunol 2009; 213(1–2): 123–30.
  161. 161. Ligocki AJ, Lovato L, Xiang D, et al. A unique antibody gene signature is prevalent in the central nervous system of patients with multiple sclerosis. J Neuroimmunol 2010; 226(1–2): 192–3.
  162. 162. Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol 2006; 59(6): 880–92.
  163. 163. Serafini B, Rosicarelli B, Magliozzi R, et al. Detection of ectopic B‐cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol (Zurich, Switzerland) 2004; 14(2): 164–74.
  164. 164. Walter BA, Valera VA, Takahashi S, et al. Evidence of antibody production in the rat cervical lymph nodes after antigen administration into the cerebrospinal fluid. Arch Histol Cytol 2006; 69(1): 37–47.
  165. 165. Arnold AC, Pepose JS, Hepler RS, et al. Retinal periphlebitis and retinitis in multiple sclerosis. I. Pathologic characteristics. Ophthalmology 1984; 91(3): 255–62.
  166. 166. Quentin CD, Reiber H. Fuchs heterochromic cyclitis: rubella virus antibodies and genome in aqueous humor. Am J Ophthalmol 2004; 138(1): 46–54.
  167. 167. Reiber H, Kruse‐Sauter H, Quentin CD. Antibody patterns vary arbitrarily between cerebrospinal fluid and aqueous humor of the individual multiple sclerosis patient: specificity‐independent pathological B cell function. J Neuroimmunol 2015; 278:247–54).
  168. 168. 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(Pt 4): 1089–104.
  169. 169. Magliozzi R, Howell OW, Reeves C, et al. A gradient of neuronal loss and meningeal inflammation in multiple sclerosis. Ann Neurol 2010; 68(4): 477–93.
  170. 170. Chaitanya GV, Omura S, Sato F, et al. Inflammation induces neuro‐lymphatic protein expression in multiple sclerosis brain neurovasculature. J Neuroinflammation 2013; 10(1): 125.
  171. 171. Gardner C, Magliozzi R, Durrenberger PF, et al. Cortical grey matter demyelination can be induced by elevated pro‐inflammatory cytokines in the subarachnoid space of MOG‐immunized rats. Brain 2013; 136(Pt 12): 3596–608.
  172. 172. Nasr IW, Reel M, Oberbarnscheidt MH, et al. Tertiary lymphoid tissues generate effector and memory T cells that lead to allograft rejection. Am J Transplant 2007; 7(5): 1071–9.
  173. 173. Thaunat O, Patey N, Caligiuri G, et al. Chronic rejection triggers the development of an aggressive intragraft immune response through recapitulation of lymphoid organogenesis. J Immunol 2010; 185(1): 717–28.
  174. 174. Thaunat O, Patey N, Morelon E, et al. Lymphoid neogenesis in chronic rejection: the murderer is in the house. Curr Opin Immunol 2006; 18(5): 576–9.
  175. 175. Columba‐Cabezas S, Griguoli M, Rosicarelli B, et al. Suppression of established experimental autoimmune encephalomyelitis and formation of meningeal lymphoid follicles by lymphotoxin β receptor‐Ig fusion protein. J Neuroimmunol 2006; 179(1–2): 76–86.
  176. 176. Kuerten S, Schickel A, Kerkloh C, et al. Tertiary lymphoid organ development coincides with determinant spreading of the myelin‐specific T cell response. Acta Neuropathol 2012; 124(6): 861–73.
  177. 177. Peters A, Pitcher LA, Sullivan JM, et al. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity 2011; 35(6): 986–96.
  178. 178. Huang Y, Caputo CR, Noordmans GA, et al. Identification of novel genes associated with renal tertiary lymphoid organ formation in aging mice. PLoS One 2014; 9(3): e91850.
  179. 179. Bonnan M. Meningeal tertiary organs: Major actors in intrathecal autoimmunity. Rev Neurol (Paris) 2014; 171(1): 65–74.
  180. 180. Lucchinetti CF, Popescu BF, Bunyan RF, et al. Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med 2011; 365(23): 2188–97.
  181. 181. Absinta M, Vuolo L, Rao A, et al. Gadolinium‐based MRI characterization of leptomeningeal inflammation in multiple sclerosis. Neurology 2015; 85(1): 18–28.
  182. 182. Aspelund A, Antila S, Proulx ST, et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med 2015; 212(7): 991–9.
  183. 183. Braga‐Neto UM, Marques ET, Jr. From functional genomics to functional immunomics: new challenges, old problems, big rewards. PLoS Comput Biol 2006; 2(7): e81.
  184. 184. Reiber H. Cerebrospinal fluid—physiology, analysis and interpretation of protein patterns for diagnosis of neurological diseases. Mult Scler 1998; 4(3): 99–107.
  185. 185. Rostasy K, Reiber H, Pohl D, et al. Chlamydia pneumoniae in children with MS: frequency and quantity of intrathecal antibodies. Neurology 2003; 61(1): 125–8.
  186. 186. Franciotta D, Di Stefano AL, Jarius S, et al. Cerebrospinal BAFF and Epstein‐Barr virus‐specific oligoclonal bands in multiple sclerosis and other inflammatory demyelinating neurological diseases. J Neuroimmunol 2011; 230(1–2): 160–3.
  187. 187. Owens GP, Burgoon MP, Devlin ME, et al. Extraction and purification of active IgG from SSPE and MS brain. J Virol Methods 1997; 68(2): 119–25.
  188. 188. Brettschneider J, Tumani H, Kiechle U, et al. IgG antibodies against measles, rubella, and varicella zoster virus predict conversion to multiple sclerosis in clinically isolated syndrome. PloS One 2009; 4(11): e7638.
  189. 189. Petereit HF, Reske D. Expansion of antibody reactivity in the cerebrospinal fluid of multiple sclerosis patients – follow‐up and clinical implications. Cerebrospinal Fluid Res 2005; 2:3.
  190. 190. Sandberg‐Wollheim M, Vandvik B, Nadj C, et al. The intrathecal immune response in the early stage of multiple sclerosis. J Neurol Sci 1987; 81(1): 45–53.
  191. 191. Fainardi E, Castellazzi M, Tamborino C, et al. Chlamydia pneumoniae‐specific intrathecal oligoclonal antibody response is predominantly detected in a subset of multiple sclerosis patients with progressive forms. J Neurovirol 2009; 15(5–6): 425–33.
  192. 192. Ahlgren C, Oden A, Bergstrom T, et al. Serum and CSF measles antibody levels increase over time in patients with multiple sclerosis or clinically isolated syndrome. J Neuroimmunol 2012; 247(1–2): 70–4.
  193. 193. Ahlgren C, Oden A, Haghighi S, et al. The effect of live, attenuated measles vaccine and measles infection on measles antibody levels in serum and CSF of patients with multiple sclerosis or clinically isolated syndrome. J Neuroimmunol 2011; 235(1–2): 98–103.
  194. 194. Bonnan M. Does disease‐irrelevant intrathecal synthesis in multiple sclerosis make sense in the light of tertiary lymphoid organs? Front Neurol 2014; 5:27.
  195. 195. Salmi A, Viljanen M, Reunanen M. Intrathecal synthesis of antibodies to diphtheria and tetanus toxoids in multiple sclerosis patients. J Neuroimmunol 1981; 1(3): 333–41.
  196. 196. Sandberg‐Wollheim M, Zweiman B, Levinson AI, et al. Humoral immune responses within the human central nervous system following systemic immunization. J Neuroimmunol 1986; 11(3): 205–14.
  197. 197. Salmi A, Reunanen M, Ilonen J, et al. Intrathecal antibody synthesis to virus antigens in multiple sclerosis. Clin Exp Immunol 1983; 52(2): 241–9.
  198. 198. Arnadottir T, Reunanen M, Salmi A. Intrathecal synthesis of virus antibodies in multiple sclerosis patients. Infect Immun 1982; 38(2): 399–407.
  199. 199. Robinson‐Agramonte M, Reiber H, Cabrera‐Gomez JA, et al. Intrathecal polyspecific immune response to neurotropic viruses in multiple sclerosis: a comparative report from Cuban patients. Acta Neurol Scand 2007; 115(5): 312–8.
  200. 200. Jacobi C, Lange P, Reiber H. Quantitation of intrathecal antibodies in cerebrospinal fluid of subacute sclerosing panencephalitis, herpes simplex encephalitis and multiple sclerosis: discrimination between microorganism‐driven and polyspecific immune response. J Neuroimmunol 2007; 187(1–2): 139–46.
  201. 201. Conrad AJ, Chiang EY, Andeen LE, et al. Quantitation of intrathecal measles virus IgG antibody synthesis rate: subacute sclerosing panencephalitis and multiple sclerosis. J Neuroimmunol 1994; 54(1–2): 99–108.
  202. 202. Otto C, Hofmann J, Finke C, et al. The fraction of varicella zoster virus‐specific antibodies among all intrathecally‐produced antibodies discriminates between patients with varicella zoster virus reactivation and multiple sclerosis. Fluids Barriers CNS 2014; 11(1): 3.
  203. 203. Otto C, Oltmann A, Stein A, et al. Intrathecal EBV antibodies are part of the polyspecific immune response in multiple sclerosis. Neurology 2011; 76(15): 1316–21.
  204. 204. Castellazzi M, Contini C, Tamborino C, et al. Epstein‐Barr virus‐specific intrathecal oligoclonal IgG production in relapsing‐remitting multiple sclerosis is limited to a subset of patients and is composed of low‐affinity antibodies. J Neuroinflammation 2014; 11:188.
  205. 205. Castellazzi M, Tamborino C, Cani A, et al. Epstein‐Barr virus‐specific antibody response in cerebrospinal fluid and serum of patients with multiple sclerosis. Mult Scler 2010; 16(7): 883–7.
  206. 206. Nociti V, Frisullo G, Marti A, et al. Epstein‐Barr virus antibodies in serum and cerebrospinal fluid from multiple sclerosis, chronic inflammatory demyelinating polyradiculoneuropathy and amyotrophic lateral sclerosis. J Neuroimmunol 2010; 225(1–2): 149–52.
  207. 207. Pohl D, Rostasy K, Jacobi C, et al. Intrathecal antibody production against Epstein‐Barr and other neurotropic viruses in pediatric and adult onset multiple sclerosis. J Neurol 2010; 257(2): 212–6.
  208. 208. Rand KH, Houck H, Denslow ND, et al. Epstein‐Barr virus nuclear antigen‐1 (EBNA‐1) associated oligoclonal bands in patients with multiple sclerosis. J Neurol Sci 2000; 173(1): 32–9.
  209. 209. Sargsyan SA, Shearer AJ, Ritchie AM, et al. Absence of Epstein‐Barr virus in the brain and CSF of patients with multiple sclerosis. Neurology 2010; 74(14): 1127–35.
  210. 210. Villegas E, Santiago O, Carrillo JA, et al. Low intrathecal immune response of anti‐EBNA‐1 antibodies and EBV DNA from multiple sclerosis patients. Diagn Microbiol Infect Dis 2011; 70(1): 85–90.
  211. 211. Virtanen JO, Wohler J, Fenton K, et al. Oligoclonal bands in multiple sclerosis reactive against two herpesviruses and association with magnetic resonance imaging findings. Mult Scler 2014; 20(1): 27–34.
  212. 212. Jaquiery E, Jilek S, Schluep M, et al. Intrathecal immune responses to EBV in early MS. Eur J Immunol 2010; 40(3): 878–87.
  213. 213. Rosengren S, Wei N, Kalunian KC, et al. Elevated autoantibody content in rheumatoid arthritis synovia with lymphoid aggregates and the effect of rituximab. Arthr Res Ther 2008; 10(5): R105.
  214. 214. Cassese G, Lindenau S, de Boer B, et al. Inflamed kidneys of NZB/W mice are a major site for the homeostasis of plasma cells. Eur J Immunol 2001; 31(9): 2726–32.
  215. 215. Corcione A, Casazza S, Ferretti E, et al. Recapitulation of B cell differentiation in the central nervous system of patients with multiple sclerosis. Proc Natl Acad Sci U S A 2004; 101(30): 11064–9.
  216. 216. Elliott C, Lindner M, Arthur A, et al. Functional identification of pathogenic autoantibody responses in patients with multiple sclerosis. Brain 2012; 135(Pt 6): 1819–33.
  217. 217. Breij ECW, Brink BP, Veerhuis R, et al. Homogeneity of active demyelinating lesions in established multiple sclerosis. Ann Neurol 2008; 63(1): 16–25.
  218. 218. Sadaba MC, Tzartos J, Paino C, et al. Axonal and oligodendrocyte‐localized IgM and IgG deposits in MS lesions. J Neuroimmunol 2012; 247(1–2): 86–94.
  219. 219. Zhou D, Srivastava R, Nessler S, et al. Identification of a pathogenic antibody response to native myelin oligodendrocyte glycoprotein in multiple sclerosis. Proc Natl Acad Sci U S A 2006; 103(50): 19057–62.
  220. 220. von Budingen HC, Harrer MD, Kuenzle S, et al. Clonally expanded plasma cells in the cerebrospinal fluid of MS patients produce myelin‐specific antibodies. Eur J Immunol 2008; 38(7): 2014–23.
  221. 221. Zhang Y, Da R‐R, Guo W, et al. Axon reactive B cells clonally expanded in the cerebrospinal fluid of patients with multiple sclerosis. J Clin Immunol 2005; 25(3): 254–64.
  222. 222. Pandey S, Alcaro MC, Scrima M, et al. Designed glucopeptides mimetics of myelin protein epitopes as synthetic probes for the detection of autoantibodies, biomarkers of multiple sclerosis. J Med Chem 2012; 55(23): 10437–47.
  223. 223. Linington C, Lassmann H. Antibody responses in chronic relapsing experimental allergic encephalomyelitis: correlation of serum demyelinating activity with antibody titre to the myelin/oligodendrocyte glycoprotein (MOG). J Neuroimmunol 1987; 17(1): 61–9.
  224. 224. Rosenbluth J, Schiff R, Liang W‐L, et al. Antibody‐mediated CNS demyelination II. Focal spinal cord lesions induced by implantation of an IgM antisulfatide‐secreting hybridoma. J Neurocytol 2003; 32(3): 265–76.
  225. 225. Blauth K, Soltys J, Matschulat A, et al. Antibodies produced by clonally expanded plasma cells in multiple sclerosis cerebrospinal fluid cause demyelination of spinal cord explants. Acta Neuropathol 2015; 130(6): 765–81.
  226. 226. Keirstead HS, Pataky DM, McGraw J, et al. In vivo immunological suppression of spinal cord myelin development. Brain Res Bull 1997; 44(6): 727–34.
  227. 227. Ramberger M, Bsteh G, Schanda K, et al. NMDA receptor antibodies: a rare association in inflammatory demyelinating diseases. Neurol Neuroimmunol Neuroinflamm 2015; 2(5): e141.
  228. 228. Niehaus A, Shi J, Grzenkowski M, et al. Patients with active relapsing‐remitting multiple sclerosis synthesize antibodies recognizing oligodendrocyte progenitor cell surface protein: implications for remyelination. Ann Neurol 2000; 48(3): 362–71.
  229. 229. Annunziata P, Cioni C, Cantalupo L, et al. Immunosuppressive monoclonal antibody to CD64 from patients with long‐term stable multiple sclerosis. J Neuroimmunol 2013; 256(1–2): 62–70.
  230. 230. Gredler V, Reindl M. B cells accumulate in the cerebrospinal fluid in inflammatory neurological diseases. J Cytol Histol 2012; S1(001): 1–7.
  231. 231. Beyer NH, Lueking A, Kowald A, et al. Investigation of autoantibody profiles for cerebrospinal fluid biomarker discovery in patients with relapsing‐remitting multiple sclerosis. J Neuroimmunol 2012; 242(1–2): 26–32.
  232. 232. Cepok S, Zhou D, Srivastava R, et al. Identification of Epstein‐Barr virus proteins as putative targets of the immune response in multiple sclerosis. J Clin Invest 2005; 115(5): 1352–60.
  233. 233. Fraussen J, Claes N, deBock L, et al. Targets of the humoral autoimmune response in multiple sclerosis. Autoimmun Rev 2014; Nov;13(11):1126-37
  234. 234. Vaughan K, Peters B, O'Connor KC, et al. A molecular view of multiple sclerosis and experimental autoimmune encephalitis: what can we learn from the epitope data? J Neuroimmunol 2014; 267(1–2): 73–85.
  235. 235. Sherer Y, Gorstein A, Fritzler MJ, et al. Autoantibody explosion in systemic lupus erythematosus: more than 100 different antibodies found in SLE patients. Semin Arthr Rheum 2004; 34(2): 501–37.
  236. 236. Rouwette M, Somers K, Govarts C, et al. Novel cerebrospinal fluid and serum autoantibody targets for clinically isolated syndrome. J Neurochem 2012; 123(4): 568–77.
  237. 237. Ligocki AJ, Rivas JR, Rounds WH, et al. A distinct class of antibodies may be an indicator of gray matter autoimmunity in early and established relapsing remitting multiple sclerosis patients. ASN Neuro 2015; 7:5.
  238. 238. Somers V, Govarts C, Somers K, et al. Autoantibody profiling in multiple sclerosis reveals novel antigenic candidates. J Immunol (Baltimore, MD: 1950) 2008; 180(6): 3957–63.
  239. 239. Lefranc D, Almeras L, Dubucquoi S, et al. Distortion of the self‐reactive IgG antibody repertoire in multiple sclerosis as a new diagnostic tool. J Immunol 2004; 172(1): 669–78.
  240. 240. Wang D, Bhat R, Sobel RA, et al. Uncovering cryptic glycan markers in multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE). Drug Dev Res 2014; 75(3): 172–88.
  241. 241. Kanter JL, Narayana S, Ho PP, et al. Lipid microarrays identify key mediators of autoimmune brain inflammation. Nat Med 2006; 12(1): 138–43.
  242. 242. Robinson WH, Fontoura P, Lee BJ, et al. Protein microarrays guide tolerizing DNA vaccine treatment of autoimmune encephalomyelitis. Nat Biotechnol 2003; 21(9): 1033–9.
  243. 243. Brennan KM, Galban‐Horcajo F, Rinaldi S, et al. Lipid arrays identify myelin‐derived lipids and lipid complexes as prominent targets for oligoclonal band antibodies in multiple sclerosis. J Neuroimmunol 2011; 238(1–2): 87–95.
  244. 244. Hecker M, Lorenz P, Steinbeck F, et al. Computational analysis of high‐density peptide microarray data with application from systemic sclerosis to multiple sclerosis. Autoimmun Rev 2012; 11(3): 180–90.
  245. 245. Wang D, Liu S, Trummer BJ, et al. Carbohydrate microarrays for the recognition of cross‐reactive molecular markers of microbes and host cells. Nat Biotechnol 2002; 20(3): 275–81.
  246. 246. Burska AN, Hunt L, Boissinot M, et al. Autoantibodies to posttranslational modifications in rheumatoid arthritis. Mediators Inflamm 2014; 2014:492873.
  247. 247. Zamvil SS, Mitchell DJ, Moore AC, et al. T‐cell epitope of the autoantigen myelin basic protein that induces encephalomyelitis. Nature 1986; 324(6094): 258–60.
  248. 248. Papini AM. The use of post‐translationally modified peptides for detection of biomarkers of immune‐mediated diseases. J Pept Sci 2009; 15(10): 621–8.
  249. 249. Brettschneider J, Jaskowski TD, Tumani H, et al. Serum anti‐GAGA4 IgM antibodies differentiate relapsing remitting and secondary progressive multiple sclerosis from primary progressive multiple sclerosis and other neurological diseases. J Neuroimmunol 2009; 217(1–2): 95–101.
  250. 250. Thiele GM, Duryee MJ, Anderson DR, et al. Malondialdehyde‐acetaldehyde adducts and anti‐malondialdehyde‐acetaldehyde antibodies in rheumatoid arthritis. Arthr Rheumatol 2015; 67(3): 645–55.
  251. 251. Boullerne AI, Rodriguez JJ, Touil T, et al. Anti‐S‐nitrosocysteine antibodies are a predictive marker for demyelination in experimental autoimmune encephalomyelitis: implications for multiple sclerosis. J Neurosci 2002; 22(1): 123–32.
  252. 252. Qin J, Goswami R, Balabanov R, et al. Oxidized phosphatidylcholine is a marker for neuroinflammation in multiple sclerosis brain. J Neurosci Res 2007; 85(5): 977–84.
  253. 253. Ho PP, Kanter JL, Johnson AM, et al. Identification of naturally occurring fatty acids of the myelin sheath that resolve neuroinflammation. Sci Transl Med 2012; 4(137): 137ra73.
  254. 254. Pasquali L, Pecori C, Chico L, et al. Relation between plasmatic and cerebrospinal fluid oxidative stress biomarkers and intrathecal Ig synthesis in multiple sclerosis patients. J Neuroimmunol 2015; 283:39–42).
  255. 255. Eggleton P, Nissim A, Ryan BJ, et al. Detection and isolation of human serum autoantibodies that recognize oxidatively modified autoantigens. Free Radic Biol Med 2013; 57:79–91.
  256. 256. Merbl Y, Kirschner MW. Protein microarrays for genome‐wide posttranslational modification analysis. Wiley Interdiscip Rev Syst Biol Med 2011; 3(3): 347–56.

Written By

Mickael Bonnan

Submitted: 12 October 2015 Reviewed: 18 March 2016 Published: 08 September 2016