Next Article in Journal
Halting Tumor Progression via Novel Non-Hydroxamate Triazole-Based Mannich Bases MMP-2/9 Inhibitors; Design, Microwave-Assisted Synthesis, and Biological Evaluation
Next Article in Special Issue
Targeting the RNA-Binding Protein HuR as Potential Thera-Peutic Approach for Neurological Disorders: Focus on Amyo-Trophic Lateral Sclerosis (ALS), Spinal Muscle Atrophy (SMA) and Multiple Sclerosis
Previous Article in Journal
Partial Decellularized Scaffold Combined with Autologous Nasal Epithelial Cell Sheet for Tracheal Tissue Engineering
Previous Article in Special Issue
Platelet Inhibition by Low-Dose Acetylsalicylic Acid Reduces Neuroinflammation in an Animal Model of Multiple Sclerosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential Biomarkers Associated with Multiple Sclerosis Pathology

by
Deepali Mathur
1,†,
Bikash Kumar Mishra
1,†,
Soumyashree Rout
1,
Francisco Jose Lopez-Iranzo
2,
Gerardo Lopez-Rodas
3,*,
Jayalakshmi Vallamkondu
4,
Ramesh Kandimalla
5,6 and
Bonaventura Casanova
7,*
1
Department of Neurology, Apollo Hospitals, Bhubaneswar 751005, India
2
Hospital of Requena, Generalitat Valenciana, 46340 Requena, Spain
3
Department of Biochemistry and Molecular Biology, Institute for Health Research INCLIVA, University of Valencia, 46010 Valencia, Spain
4
National Institute of Technology, Warangal 506004, India
5
Department of Biochemistry, Kakatiya Medical College, Warangal 506007, India
6
Department of Applied Biology, Indian Institute of Chemical Technology (CSIR-IICT), Hyderabad 500007, India
7
Unitat de Neuroimmunologia, Hospital Universitari i Politècnic La Fe, la Universitat de València, 46026 Valencia, Spain
*
Authors to whom correspondence should be addressed.
Deepali Mathur and Bikash Kumar Mishra are equal authors.
Int. J. Mol. Sci. 2021, 22(19), 10323; https://doi.org/10.3390/ijms221910323
Submission received: 3 August 2021 / Revised: 21 September 2021 / Accepted: 23 September 2021 / Published: 25 September 2021

Abstract

:
Multiple sclerosis (MS) is a complex disease of the central nervous system (CNS) that involves an intricate and aberrant interaction of immune cells leading to inflammation, demyelination, and neurodegeneration. Due to the heterogeneity of clinical subtypes, their diagnosis becomes challenging and the best treatment cannot be easily provided to patients. Biomarkers have been used to simplify the diagnosis and prognosis of MS, as well as to evaluate the results of clinical treatments. In recent years, research on biomarkers has advanced rapidly due to their ability to be easily and promptly measured, their specificity, and their reproducibility. Biomarkers are classified into several categories depending on whether they address personal or predictive susceptibility, diagnosis, prognosis, disease activity, or response to treatment in different clinical courses of MS. The identified members indicate a variety of pathological processes of MS, such as neuroaxonal damage, gliosis, demyelination, progression of disability, and remyelination, among others. The present review analyzes biomarkers in cerebrospinal fluid (CSF) and blood serum, the most promising imaging biomarkers used in clinical practice. Furthermore, it aims to shed light on the criteria and challenges that a biomarker must face to be considered as a standard in daily clinical practice.

1. Introduction

Multiple sclerosis (MS) is a chronic, inflammatory, and autoimmune disease of the central nervous system (CNS) that causes damage to myelin sheaths and axons. It affects more than two million people worldwide and 5–20 per 100,000 persons in India [1,2]. Young individuals aged between 20 and 40 years are mainly affected, although MS also develops in children, teenagers, and the elderly [3]. According to Lubin et al.’s classification, MS is categorized into relapsing and progressive stages, with disease activity and/or progression [4]. Ninety percent of patients are affected with the relapsing remitting clinical form of MS (RRMS). The majority of these patients eventually transition into secondary progressive MS (SPMS), which causes further deterioration and neurological disability [5]. The progressive form of MS is comprised of primary progressive MS (PPMS) and SPMS. Approximately 15% of patients are affected with PPMS, which is considered the subtype with the worst prognosis [6]. Due to heterogeneous clinical presentation of MS, there is no specific ‘diagnostic test’ available in the laboratory, leading to delays in diagnosis and determining a prognosis. Therefore, accurate diagnostic tools are required to understand the development and progression of disease, and the use of blood and cerebrospinal fluid (CSF) biomarkers would aid in the diagnosis, prognosis, and monitoring responses to current treatments [7]. The later objective would be of particular interest in SPMS, since most of the patients that have this MS-subtype do not respond well to new disease-modifying therapies (DMT) used to treat MS [8,9].
Neuromyelitis optica spectrum disorder (NMOSD) is historically considered to be the Asian form of the optic-spinal form of MS. Initially, NMO was considered as an isoform of MS and not as a distinct entity of autoimmune and demyelinating CNS disease. However, the discovery of NMO-specific IgG antibodies found in the serum of NMO patients, mainly against aquaporin 4 protein and expressed in astrocytes, allowed the differentiation of MS from NMO patients. Moreover, extensive clinic research and the development of highly sensitive and specific methods to assess myelin oligodendrocyte glycoprotein antibody (MOG-Ab) disease allowed the diagnosis of a group of patients with MOG antibodies, which are also clinically and phenotypically different from MS or NMO. Therefore, MOG-Ab disease is now viewed as a separate entity that requires a specific, differentiated treatment. MOG-Ab disease is also detected in patients with acquired demyelinating syndrome [10,11,12], clinically isolated syndrome, optic neuritis, transverse myelitis, NMO spectrum disorders, and MS [13,14].
Therefore, the aforementioned data clearly justify a detailed investigation of biomarkers to better understand the factors that contribute to the development of MS, its exacerbation into the various subtypes, the effects of treatment, and the disease prognosis. Biomarkers would also be useful in differential diagnoses between MS and other CNS demyelinating diseases with similar signs or symptoms.
In recent years, several remarkable reviews have been published that highlight the importance of biomarkers in the prediction, diagnosis, and outcome of treatments in MS e.g., [15,16,17,18]. In this physically and mentally disabling disease, being able to make relevant information on the potential best biomarkers easily accessible to MS-clinicians is essential for daily practice. Bearing this in mind, the objective of this review is to describe and analyze the main biomarkers clinically available, along with their present and potential future uses in MS patients.

2. The Criterion to Be Considered as a Standard Biomarker

The characteristics used to classify a biomarker as ideal are as follows:
(1)
It must have the ability to differentiate between a patient and a healthy individual;
(2)
It must be expressed at an early stage before the disease progresses;
(3)
It must be easy to evaluate, safe for patients, and informative for subsequent management of the disease;
(4)
It must offer reproducible results. Furthermore, the biomarker must be sensitive, specific, and must have a clear predictive value.
In this way, the identification of specific biomarkers can help to diagnose the disease, understand the progression of the disease, and what the response to therapeutic regimens would be.
In MS, body fluids such as blood and CSF have different merits and demerits for use in identifying the specific biomarkers they contain. The detection of biomarkers in a blood sample is a safe, fast, and simple technique, and, contrasting CSF, biomarkers can be measured at different time points without undue inconvenience to patients. However, there are certain demerits to the blood sample method. For instance:
(1)
The measurement of biomarkers in a blood sample may not necessarily reflect changes in the CNS, unlike CSF;
(2)
In blood samples, markers are affected by many clinical and biological processes;
(3)
The concentration of biomarkers in blood may be lower than the corresponding amount in the CSF.
Although measuring a biomarker in the CSF directly has the advantage of reflecting changes in the CNS, these samples must be obtained by lumbar puncture, which is a difficult and traumatic process that cannot be repeated with the frequency of a simple blood sample.

3. Challenges in the Development of Biomarkers

There is no doubt that there are several obstacles that must be overcome in order to establish biomarkers as ideal for clinical use. For instance, there are different detection strategies and kits available on the market to detect molecular biomarkers. However, even small variations in the available detection methods can result in significant differences for the same tested biomarker, and thus seriously influence the significance value of the biomarker. For example, for interleukin IL-21, two different ELISA kits were used to test its potential for possible use as a biomarker, and they found significantly different results [19,20]. It is therefore essential to employ different detection methods to confirm the potential validity of a biomarker.
Likewise, the results can vary significantly when the validity of a biomarker is established in a small population and subsequently applied in a large independent cohort. For instance, studies with a small number of patients showed that anti-myelin oligodendrocyte glycoprotein (MOG), anti-myelin basic protein (MBP), or anti-KIR4.1 antibodies could serve as a predictor of the development of MS after a first demyelinating event [21,22]. However, subsequent studies with large population cohorts could not confirm this prediction [23,24]. Although there are other challenges that potential new biomarkers face, the robustness of the assay used and its application in large populations must be taken into account when developing them, in order to minimize the error of their use in patients.

4. Classification of Biomarkers

Biomarkers are classified according to:
(A)
Personal susceptibility or predictiveness;
(B)
Diagnosis;
(C)
Prognosis;
(D)
Disease-associated activity;
(E)
Response to treatment in different disease courses of MS (Table 1).
In this review, we focus primarily on the first four criteria, given that responses to clinical treatments frequently use biomarkers from these other groups, some examples of which will be discussed throughout this review.

4.1. Predictive Biomarkers

The predictive biomarkers are useful for identifying particular individuals who are at risk for developing MS. People who are at an increased risk of developing MS and would benefit from targeted screening include:
(A)
Children and siblings of MS patients;
(B)
Healthy individuals diagnosed with clinically isolated syndrome (CIS) and other disorders related to the nervous system.
These risk groups were determined by identifying risk factors that correlate with MS [25]. The immune reaction to the Epstein–Barr virus (EBV) or human herpes virus (HHV)-6, or the presence of anti-myelin oligodendrocyte glycoprotein and anti-myelin basic protein (MBP) antibodies are good examples of predictive biomarkers (Table 2).
  • Epstein–Barr virus (EBV)
There is considerable experimental evidence suggesting an association between EBV and MS due to the similarity in the geographical distribution of the appearance of both diseases, the existence of a greater number of reported cases of infectious mononucleosis in MS patients, and higher titers of specific EBV-associated antibodies in patients with an increased risk of MS [26]. Abrahamyan et al. studied the prevalence of Epstein–Barr nuclear antigen (EBNA)-1 and viral capsid antigen (VCA) antibodies in serum from a cohort of 901 patients with CIS or early RRMS [27]. The results showed that 100% of the patients with CIS/RRMS were EBV-seropositive, thus suggesting that a negative EBV serology in patients with suspected inflammatory disease of the CNS should alert clinicians to consider diagnoses other than MS. In addition, in the largest population of EBV-CIS evaluated to date (a cohort of 1047 cases of CIS), only one was seronegative for EBNA-1, demonstrating that, while it is possible to be truly seronegative for EBV and develop MS, it is extremely rare [28].
  • Human herpesvirus type-6 (HHV-6)
The experimental results show high levels of the expression of the HHV-6 virus in oligodendrocytes close to the MS plaques [65], suggesting that there is a definitive causal relationship between HHV-6 infection and the development of MS [66]. Evidence of HHV-6 neurotropism was also found in MS from viral DNA found in the brain and CSF of MS patients. Furthermore, in clinical case-control studies, increased expression of HHV-6 genes, with higher levels of virus mRNA and proteins, were also found in the oligodendrocytes of MS patients [67,68,69]. In a more recent work, Dominguez-Mozo et al. described that higher titer of IgG HHV-6 antibodies are related to the occurrence of disease relapses in patients with MS and that treatment with natalizumab drastically reduces both the relapse rate and IgG HHV-6 antibody titers. Based on these data, the authors highlight the potential role of these antibodies not only as predictive factors, but also as early biomarkers of drug response in patients with MS [70].
  • Anti-MOG and anti-MBP antibodies
Oligodendrocytes are the myelin-forming cells of the CNS, and therefore any cellular damage to oligodendrocytes, for instance by autoantibodies, can lead to the loss of myelin sheath structure. The presence of antibodies against myelin proteins in the serum reflects an autoimmune attack against CNS myelin. Thus, anti-myelin autoantibodies, such as MOG and MBP, in the serum of patients with CIS can be considered as predictive biomarkers of disease. Berger et al. found antibodies against MBP in the serum of CIS patients who developed MS [21], and pointed out that the presence of anti-MBP antibodies in childhood increases the risk of demyelinating encephalomyelitis [112]. Subsequent studies showed that MS patients with anti-MOG antibodies are at an increased risk of developing MS and have a higher relapse rate [113]. Despite these results, the studies are not entirely conclusive since, for instance, Kuhle et al. observed that a clear connection between anti-MOG and anti-MBP presence and CIS to MS conversion is not apparent. For instance, Kuhle et al. observed that a clear connection between anti-MOG and anti-MBP presence and CIS to MS conversion is not apparent [114]. Furthermore, Kuerten et al. used microarrays to analyze antibodies against 205 myelin antigens in a cohort of 13 MS patients. Microarray significance analysis identified a subset of 64 myelin antigens, including MOG and MBP, for which widely elevated levels of anti-myelin autoantibodies could be detected in the plasma of MS patients. Despite this, the authors noted that the levels of significance were not high enough to serve these antibodies as a good predictive clinical biomarker for MS [114]. Based on these results, and those obtained by other authors (reviewed in [115]), anti-MOG antibodies would not be adequate biomarkers for the diagnosis or prognosis of MS, but rather for its differential diagnosis with MOG+-CNS demyelinating disease representing a new distinct disease entity.

4.2. Diagnostic Biomarkers

The purpose of diagnostic biomarkers is to confirm that a patient has a certain pathology, such as MS, and thus allow clinicians to discriminate between them and healthy people or those suffering from another potentially related disorder (Table 2 and Table 3). For MS, the diagnosis of the first clinical events of relapsing-remitting MS with disseminated inflammation in space can be confirmed by magnetic resonance imaging and by the existence of oligoclonal immunoglobulin bands detected in CSF. This modified McDonald MS diagnostic criteria include oligoclonal bands replacing the classic criteria of diffusion over time. Thus, when published MS data were compared by applying classic 2010 MS diagnostic criteria or 2017 modified McDonald MS diagnostic criteria, it was found that around 37% of the patients were correctly diagnosed with the 2010 criteria, and surprisingly, the number increased to 68% when the 2017 criteria were applied [174]. Therefore, the recent criteria, which include the existence of detected oligoclonal bands in CSF, provides a faster and more cost-effective approach to the diagnosis of MS than the classical criteria. However, the McDonald’s 2017 criteria have limitations when they are applied to patients with an atypical clinical syndrome or other inflammatory disorders of the CNS. These patients require expert MS clinicians for an accurate diagnosis of the disease.

4.2.1. MRI as Diagnostic Biomarker

Magnetic resonance imaging (MRI) is probably the single most important tool for diagnosing MS. This technique makes it possible to determine localized lesions in SNC. In MS, if lesions are found in the white matter of the brain, this indicates that MS is developing from CIS. The MRI study for MS includes:
(A)
T1-weighted or longitudinal magnetization relaxation time;
(B)
T2-weighted or transverse magnetization relaxation time;
(C)
A post-contrast scan. T1-weighted lesions are used primarily to detect any abnormalities in the integrity blood-brain barrier (BBB). Hypointense T1 lesions (also referred to as black holes) are used as a marker representing the loss of axons that occur during the development of MS.
MR imaging is classified into conventional and unconventional techniques (Table 3) and Figure 1 shows clinical examples of these techniques used in MS patients.

Conventional Image Techniques

  • T2- weighted MR imaging
MS lesions are typically detected on T2-weighted MRI images. T2-weighted hyperintense lesions have considerable diagnostic importance as they involve multiple clinical mechanisms of MS, such as axon loss, demyelination, and inflammation and oedema in the CNS. Through this method, both white matter (WM) and gray matter (GM) lesions can be identified. The WM and GM lesions indicate whether there is focal and cortical demyelination, and also whether there is significant axonal loss. Therefore, it is undoubtedly the most important imaging biomarker for studying neurodegeneration during MS [29].
  • T1-weighted images with gadolinium enhancement
T1-weighted lesions are used to detect BBB dysfunction in MS. T1-weighted images also provide data on CNS neurodegeneration by analyzing black holes and atrophy. Black holes describe axonal damage and destruction of neuronal tissue, while atrophy shows axonal loss that would likely occur through tissue damage within the lesions. Quantifying the number and measuring the volume of T1 black holes would correlate well with MS disability, which is why it has been proposed in the literature as a potential biomarker for CNS neurodegeneration [30,31]. MRI with Gd enhancement is useful for analyzing active inflammation, since Gd would easily enter the CNS through BBB damage [32,33].
  • Fluid Attenuated Inversion Recovery Sequence (FLAIR)
Due to the presence of excessive water and a high spin density in MS lesions, sometimes it is not possible to get a better figure of parenchymal changes in MS development. To avoid these difficulties, the FLAIR sequence technique suppresses the resonant signals of water, enhancing the hyperintense signals of the MS lesions. In this approach, intraparenchymal changes can be detected in MS, especially in the cortical and juxtacortical areas [34,35]. The technique is not sensitive enough to detect lesions in the periventricular region, which are usually detected using the T2-FLAIR sequence [34].
  • Proton density (PD)-weighted spin-echo (SE) images
Long repetition time (TR) and shorter echo form PD-weighted (proton density weighted) image forms are also well-known image biomarkers. Eco sequences were used earlier in the form of a fast spin-echo (FSE). Dual and multi-echo sequences can also be used to get PD and T2 weighted images. According to Chong in 2016, PD-FSE exposes larger lesions, which appears as numerous smaller lesions in T2- FSE images. Additionally, 32% more cervical cord lesions are identified through this procedure than through T2- FSE images, thus providing more information about neurodegeneration [36].
Images with a long repetition time (TR), shorter echo PD-weighted (proton density weighted) or, previously, fast spin echo (FSE) images are used as a useful technique in MS clinic. To obtain PD- and T2-weighted images, the more sensitive dual and multi-echo sequences can also be used. As highlighted by Chong et al., PD-FSE exposes larger lesions found in the form of numerous smaller lesions on T2-FSE images. By this procedure, 32% more extra cervical cord lesions are identified than with T2-FSE imaging, although the latter provides more information on neurodegeneration in the CNS [36].

Non-Conventional Image Techniques

  • The Magnetization Transfer Imaging (MTI)
MTI is a type of MRI technique that is applied to expose the interaction between protons that are present in three states:
(a)
Bound to macromolecules;
(b)
In free water;
(c)
As water in the hydration layer between macromolecules and free water.
MTI detects and differentiates the specific lesions that define MS to determine the severity of the disease and to describe the pathological changes that occur during MS development [35,37].
MTR is a sensitive method by which the pathogenesis of MS of myelinated white matter lesions can be assessed relative to the intact white matter, such that the decrease in the MTR value is correlated with both demyelination and axonal loss [38,39]. MTR can also provide information on the degree of optic nerve demyelination during optic neuritis. The MTR value of the optic nerve decreases, which is correlated with the thickness of the retinal nerve fiber layer (RNFL) and axonal loss, making it relevant for evaluating pathological and paraclinical visual changes during the disease [40].
  • Diffusion-Weighted (DWI) and Diffusion Tensor (DTI) Images
DWI is another MRI technique for evaluating Brownian motion of H2O. This technique studies the anatomy and potential pathologies affecting white matter (NAWM). The DWI estimates the so-called apparent diffusion coefficient (ADC) of water in the brain, which increases if there are alterations in cell structure and white matter tracts [41]. The technique is capable of distinguishing between different pathologies that affect the CNS, such as ischemia, infection, tumors, neurodegeneration, etc., by calculating the microscopic movement of water molecules whose diffusion is atypical in EM plaques [42]. Although this technique is useful for analyzing ischemic cerebrovascular accidents, its status as a marker of MS is not well established and it is used less than other MRI techniques. For instance, DWI is unable to measure the volume of tissue loss in MS lesions and cannot reveal modifications generated by other pathologies that occurred in white matter.
DTI is an MRI refinement method that evaluates the three-dimensional diffusion of water in multiple directions in space. DTI provides much more information on the pathogenesis of MS compared to T1- or T2-weighted MRI techniques. In DTI, several physical parameters are evaluated, such as:
(a)
Axial diffusivity (DA)
(b)
Radial diffusivity (RD);
(c)
Mean diffusivity (MD);
(d)
Fractional anisotropy (AF) [42].
The DA assesses the loss of axons while the RD is associated with the status of the myelin layer. In the early stages of MS, the DA value is low in normal-appearing white matter (NAWM), increasing in the later stages of MS [43]. On the other hand, DTI uses the failure analysis (FA) process to reveal the global direction of water diffusion, which increases in white matter tracts and decreases in fibers disorganized by MS [44].
The evaluated DTI values serve as biomarkers for the diagnosis and evolution of MS. MD has been established as a useful and predictive biomarker for estimating the onset or relapse of MS, since it is sensitive to changes in the tissues that precede the breakdown of the blood–brain barrier (BBB) for at least 5 months and can predict injury after restoration of the BBB [45]. DA is used as a potential biomarker of axonal degeneration and RD is used to determine the degree of demyelination over the course of MS [46,47,48].
  • Magnetic Resonance Spectroscopy (MRS)
Nuclear magnetic resonance spectroscopy (MRS) is a non-invasive technique that estimates the cellular metabolism in the CNS. The MRS measures the biochemical levels of molecules such as N-acetylaspartate, choline, creatine, glutamate, and glutamine in various pathologies, along with their deviation from natural physiological levels, as occurs in the pathogenesis of MS. Specifically, a decrease in the level of N-acetylaspartate is considered a marker of neuronal/axonal loss, while choline estimates the increase in rotational cell membrane components that is observed during demyelination or gliosis. For its part, glutamate is used as a biomarker of acute inflammation, a reduced level of N-methyl aspartate indicates a decrease in oedema in MS lesions, and GABA is decreased in the SPMS form of MS [49,50].
  • Optical Coherence Tomography (OCT)
Optical coherence tomography (OCT) is used to take images of axons and the unmyelinated axonal layers of the retina (RNFL). The variation of the RNFL reflects neurodegeneration and oedema in MS patients. Lower values of RNFL are considered a high-resolution, objective, non-invasive, and easily quantifiable in vivo biomarker of MS and represent the axon loss associated with cerebral atrophy [51,52].
  • Positron Emission Tomography (PET)
Positron emission tomography (PET) is an imaging technique used to estimate cell and tissue metabolism through the in vivo incorporation of a radiopharmaceutical. The heterogeneity of MS lesions, and the changes in inflammation of the NAWM and GM in the course of MS, can be obtained through PET images. The focal point of this technique is to produce functional images of the brain’s innate immune system, that is, of the microglia and macrophages. For instance, activated microglia secrete oxygen radical species (ROS) and pro-inflammatory cytokines in response to the neural injury that causes the inflammation characteristic of MS. Therefore, the PET technique is used as a biomarker and shows the process of neurodegeneration and neuroinflammation in the CNS of patients with MS [53,54].

4.2.2. Diagnostic Biomarkers in CSF and Serum

  • Oligoclonal bands (IgG) and (IgM)
Oligoclonal bands (OCB) are immunoglobulins (Ig) produced intrathecally and, although they are considered an immunological characteristic of MS, they are not exclusive to MS, as they are also found in other CNS pathologies [71,72,73]. OCBs are present in the CSF of more than 95% of MS patients, are absent in serum, and serve as an important criterion for the diagnosis of MS (Table 3). Among the oligoclonal bands, those of the IgG type (OCGB), produced by B cells, are possibly the most important biomarker attributed to CNS demyelinating spectrum disorders. The OCGB serves as a diagnostic element in patients with Clinically Isolated Syndrome (CIS), and is also associated with the development of Clinically Defined MS (CDMS), due to the presence of a higher level of IgG in the CSF of these patients. The presence of OCGB is also essential to predict the progression of Radiologically Isolated Syndrome (RIS) patients to CIS and from CIS to MS [74,75], whose sensitivity and specificity is 88% and 86%, respectively [76]. The presence of high levels of OCGB in the CSF of patients with CIS favors the transition to CDMS [77,78] and the subsequent progression of patients to definitive MS [79].
The presence of OCGB is also useful in predicting Optic Neuritis (ON) in MS [80,81], although they are not able to report the intensity of a second relapse. Several lines of evidence indicate that patients with OCGB present in their CSF exhibit a higher level of inflammatory activity, which leads to significant tissue damage [82,83,84], a greater degree of lesions, and greater brain atrophy [85,86,87,88].
  • Immunoglobulin G index (IgG index)
The levels of IgG and IgM OCBs released intrathecally in MS patients indicate the clonal extension of B cells and plasma cells in the CNS. The relative amount of IgG in the CSF, compared to that present in serum, is evaluated by the IgG index. The IgG index is estimated as the ratio of IgG to albumin in CSF compared to the ratio of IgG to albumin in serum [89]. On the other hand, the albumin quotient (albumin in CSF/albumin in serum) indicates the alteration to the integrity of the blood–brain barrier in MS [90]. It has been estimated that a patient may be diagnosed as having MS when the IgG index exceeds the value of 0.7, which means an increased synthesis of intrathecal IgG antibodies in the CNS which in turns triggers the symptoms of MS. Thus, the IgG index serves as an important biomarker for the diagnosis of MS and is routinely determined during the MS diagnostic.
  • Kappa Free (KFLC) and Lambda Free Light Chains (LFLC)
Plasma cells produce KFLC and LFLC during antibody synthesis, and these proteins can be detected in both serum and CSF of patients [91]. Presslauer et al. found that MS patients have a higher amount of KFLC secreted in their CSF and in their blood serum [92]. Rinker et al. demonstrated a clear correlation between the presence of an increasing amount of KFLC and the occurrence of future disabilities in MS patients [93]. Moreover, Villar et al. found that KFLC levels increase in the CSF of CIS patients and that these altered levels lead to the conversion of CIS to CDMS, suggesting its use as a marker of MS progression [94]. It has also been proposed that the LFLC subunit can be used as a predictive biomarker for intrathecally produced immunoglobulins in inflammatory disorders of the CNS [116].
  • Anti-Aquaporin-4 (AQP4) antibodies
Aquaporin-4 (AQP4) is expressed in CNS astrocytes and its function is to transport water through the cell membrane and maintain the homeostatic balance within the CNS. MS patients lack the expression of this protein, while 38–75% of patients with Neuromyelitis Optica (NMO) have AQP4 antibodies [117]. Since NMO is a rare disease in which the immune system attacks myelin surrounding the optic nerve and spinal cord, the differentiation between NMO and MS is challenging due to their similar clinical features. Patients with NMO have specific IgG antibodies in their serum directed against AQP4, which is expressed in astrocytes. Therefore, the specific immunoreactivity of this biomarker helps to differentiate between patients with NMO and MS, and also improves the determination of other disorders related to immunity also affecting the CNS [154,155].

4.3. Biomarkers of Prognosis

Prognostic biomarkers are used to predict the potential response of a patient to treatment in terms of efficacy and/or safety, which allows clinicians to make the most appropriate clinical and therapeutic decisions.
  • Oligoclonal bands (IgM)
The IgM-type immunoglobulins (OCMBs) were shown to correlate with MS activity [156] and predict an aggressive course in patients with RRMS during the early stages of the disease [157,158,171,172]. The presence of OCMB is associated with an increase in retinal axonal loss in MS [171], the thinning of the retinal nerve fibre layer [172], and its deposition influences inflammatory processes in the brain, generating greater lesions in the CNS [173].
  • Chitinase-3-Like-1 precursor (CHI3L1)
Several lines of evidence indicate that the levels of the extracellular glycoprotein CHI3L1 are found to be elevated in CSF of patients with inflammatory diseases of the CNS. This biomarker is expressed in astroglia, in WM plaques, in NAWM, and in the brain lesions of MS patients [55]. Canto et al. found elevated CSF CHI3L1 levels that correlate with the rapid development of disability during MS progression. CHI3L1 can be used as a biomarker in the transformation of CIS into CDMS [56], being elevated in patients with RRMS and SPMS compared to healthy individuals [57]. Likewise, higher levels of CSF CHI3L1 are found in T1 and T2 lesions and in the brain parenchymal fraction of MS patients. Therefore, it is suggested that CHI3L1 could be another promising prognostic biomarker of MS.
  • Neurofilaments (NF)
Neurofilaments (NFs) belong to type IV intermediate filaments and shape neurons. In the CNS they are abundant in the cytoplasm of neurons and are composed of four subfilaments of different molecular weights:
(a)
NF-L (polypeptide light chain) of 68 kDa;
(b)
NF-M (neurofilament of medium size) of 150 kDa;
(c)
NF-H (heavy chain) from 190 to 210 kDa and α-internexin [95].
If axonal damage occurs in the CNS, the filaments are secreted into CSF or blood serum [96]. The NF-L light chain can be considered as a prognostic biomarker that provides information on the transformation of CIS into RRMS [97,98]. Likewise, a high level of NF-L in CSF is considered a predictive marker of disease severity and progression to SPMS [99], and of disability and cognitive impairment during the conversion of patients to CDMS [100,101]. On the other hand, Gunnaarsson et al. observed that NF-L is associated with CNS damage, so it can also be used as a neurodegeneration biomarker in MS [102]. Kuhle et al. further demonstrated that NF-L is a marker of tissue damage and disease activity in patients with RRMS, suggesting that it is also a prognostic biomarker [103]. Patients with RRMS and SPMS have a high concentration of NF-H, and this NF is used as a prognostic marker in the development of MS and the future disability of patients [104,105].
  • miRNAs
Non-coding micro RNA molecules (miRNAs) can be detected in both serum and CSF samples. Although mRNAs are highly unstable, the miRNAs are relatively highly stable in biological fluids. Many studies of miRNA have revealed that alteration in the level of some miRNAs is related to the conversion of CIS to MS [118]. For instance, a higher level of miRNA-922 is found in patients with CIS and is associated with increased transformation of CIS into RRMS [119]. In another study, Bergmann et al. found that miRNA-150 present in plasma can function as a prognostic marker, since it influences in the conversion of CIS to MS [120]. Furthermore, Ma et al. showed that miR-19a, miR-21, miR-22, miR-142-3p, miR-146a, miR-146b, miR-155, miR-210, and miR-326 are up-regulated in MS [121]. In contrast, miR-15a, miR-15b, miR-181c, and miR-328 are downregulated in MS patients [121]. Therefore, miRNAs are being considered as promising prognostic biomarkers in patients with MS, as they are able to adequately predict the conversion of patients with CIS to MS.

4.4. Biomarkers of Dysfunction and Pathology

MS-type neurological disease activity is defined as the appearance of new neurological symptoms, the recurrence of a previous condition, identifiable radiological activity, or the progression of disability. Clinicians assign a score that predicts whether symptoms have resumed or ceased and whether it is necessary to continue or modify medication to control the disease.

4.4.1. Biomarkers of Immunological Dysfunction

  • Cytokines
Neuroinflammation occurs during the relapsing phase of MS, releasing numerous cytokines and chemokines into the CSF, which in turn produces CNS lesions characteristic of MS. Many articles have been published in the literature that describes the changes in certain cytokines during the course of MS, and that are detectable in the CSF of patients [58,59,60,61,62], or in their serum [62,63,64,65]. Various authors have proposed cytokines as potential biomarkers of immunological dysfunction in MS.
The chemokine CXCL13, interacts with the CXCR5 receptor, and results in the activation of B and T helper cells in demyelination lesions. Furthermore, a higher level of CXCL13 is found to be associated with the conversion of CIS to MS [58]. Mouzaki et al. found that, through cytokines, it is easy to differentiate MS patients from other inflammatory CNS disorders [59]. Kim et al. also showed that any imbalance in the IL-1 signaling leads to CNS demyelination [60]. Huang et al. investigated protein biomarkers in CSF and plasma using a highly sensitive proteomic immunoassay. The cases from two independent cohorts were compared with healthy controls and patients with other neurological diseases. Ten up-regulated proteins were identified in CSF, including CCL11, IL-12B, CD5, MIP-1a, and CXCL9 in MS patients [61]. Moreover, CCL11 was associated with disease duration, particularly in patients with SPMS subtype [61]. Bai et al., in a meta-analysis of 226 studies with 13,526 MS patients and 8428 healthy controls, showed that 13 CSF cytokines are significantly associated with MS, and among them, CCL21, IL-15, CCL19, CCL11, CCL3, and CXCL13 showed larger standardized mean differences (measured as ES parameter or effective sizes differences between groups with statistical significance) in cytokine concentrations between MS patients and healthy controls [62].
Cytokines have also been proposed as potential biomarkers in blood serum, a clearing fluid that is easier and less traumatic to obtain, which is why it would be highly useful in daily clinical practice. For instance, the relapse rate and prevalence of MS patients was correlated with the levels of interleukin IL-6 in their blood serum [63] and any imbalance in IL-1 signaling leads to increased CNS demyelination [64]. Mouzaki et al. pointed out that measuring some cytokines in serum may allow clinicians to differentiate MS patients from other CNS inflammatory disorders at an early stage [59]. In the same meta-analysis by Bai et al., of the 37 serum cytokines analyzed in the meta-analysis, 21 cytokines were significantly associated with MS. Among these, CCL20, IL-23, IL-21, IL-12p40, IL17F, IL22, and IL2R had large ES values which differentiated between MS patients and healthy controls [62].

4.4.2. Biomarkers of Demyelination

  • Myelin basic protein (MBP)
Although the levels of the MBP protein increase in the CSF of patients with MS during acute demyelination, they are not considered a good prognostic biomarker [106,107]. This is because MBPs in CSF tend to remyelinate demyelinating lesions, but the reverse pathway has not been yet demonstrated [108,109]. The literature reveals that a higher level of MBP has been found in the CSF of MS patients [110] and that MBP levels increase dramatically during the relapse of MS patients [111]. Despite this, it is not considered a good pathology biomarker, given the inconsistency of the results obtained.

4.4.3. Biomarkers of Axonal Damage

  • Neurofilaments (NF)
MS patients with high levels of axonal degeneration and neuronal death have a higher level of neurofilament type NF-H in the progressive clinical course of MS [122,123]. Likewise, higher levels of NF-H have been found in patients with CIS and RRMS [124], and correlate with the relapse activity of patients with CIS and RRMS [125]. For its part, the NF-L type, after dissociation, disperses from the parenchyma into the CSF due to its low molecular mass and hypophosphorylation [125], and a high level is also found in patients with MS or CIS [126,127]. It has even been shown that NF-L levels increase in the CSF during acute MS relapse and in MS patients with higher relapse rates. Finally, it was reported that in cases of transition from RRMS to SPMS, a higher level of NF-L is also found [97]. Therefore, neurofilaments are good candidates to be used as a prognostic biomarker to determine axonal damage and estimate the efficacy of MS treatment.
  • Microtubule-associated protein Tau
Tau protein can be used as a biomarker of axonal loss in patients with MS [128]. When the abnormal phosphorylation of the protein occurs, the microtubules are unstable, generating insoluble tau that acts as a neurotoxic agent. Neurotoxic tau results in the development of common neurodegenerative diseases such as MS [129,130]. Following neuronal injury, tau protein is released into serum and CSF [131], abnormally hyperphosphorylated and in an insoluble tau form, can be found in patients with SPMS and PPMS, leading to the progression of the disease [132,133]. Patients with CIS show elevated levels of both tau and NF-L, which favors the transformation of CIS into CDMS, and the progression of MS [124,133].
  • N-acetylaspartate (NAA)
Studies have revealed that reduced levels of NAA are found in MS lesions, surrounding NAWM and cortical grey matter [134]. Disease progression in MS and disability are associated with a low level of NAA [128]. Other studies have shown that a decreased level of NAA is present in CIS patient’s grey matter and in some white matter lesions [135,136]. Narayanan et al. found that MS patients who are treated with interferon-β 1b for one year express a higher concentration of NAA [137].
  • Amyloid-precursor protein (APP)
Although the accumulation of protein Aβ is seen primarily in Alzheimer’s disease (AD), recent studies have indicated a link between amyloid-β and MS. Increasing evidence suggests that myelin damage results in proteolytic processing of APP. In damaged axons, the cytoskeleton causes disruption of axoplasmic flow, possibly due to a massive influx of calcium across the membrane [138,139]. Changes in axoplasmic flow lead to the deposition of APP in the axons, indicating a failure of axonal transport. Mattson et al. found that the level of α-Sapp and β-Sapp decreased in the CSF of MS patients compared to healthy individuals [140]. APP-positive axons in MS patients are associated with the development of CNS lesions [141]. Therefore, this protein is being used as a potential biomarker to determine the progression of MS. APP is not only found in reactive glial cells during demyelination, but is also expressed during remyelination, with APP being produced by astrocytes during demyelination [142]. The findings of Gehrmann et al. reveal that a higher concentration of APP is detectable in MS patients compared to healthy individuals [141].
  • Ionic Imbalance
In myelinated axons, sodium channels are located in the nodes of Ranvier, while in demyelinated axons, they are distributed along the axons in an ATP-dependent process. This increased demand for ATP leads to the failure of Na+/K+ ATPase, caused by the presence of immune infiltrates in the CSF of MS patients, which leads to an intracellular increase in the ionic gradient of Na+. The increased level of intracellular Na+ reverses the activity of the Na+/Ca2+ exchanger, resulting in the increase in Ca2+ in the axoplasm, and finally in axonal damage and mitochondrial dysfunction [143,144]. Our research group has found that patients with RRMS with IgG and IgM antibodies (IgG+/IgM+) have less energy to repair axonal lesions, due to repression of genes related to ATP production, so they have more aggressive clinical symptoms and greater inflammation compared to IgG+/IgM patients [145]. Oligodendrocyte progenitor cells (OPC) in MS patients with IgG+/IgM+ RRMS decrease their ability to repair demyelinated regions, due of lack of sufficient energy to repair neuronal damage, which leads to a poor clinical prognosis in contrast to IgG+/IgM RRMS patients [145,146]. Thus, energy failure and toxic sodium accumulation may initiate a vicious cycle, in which the increased intracellular Na+ concentrations may provoke reverse action of the Na+/Ca2+ exchanger and thus cause Ca2+ accumulation, leading to a cellular ion imbalance which increases activation of neurodegenerative signaling cascades [147].
It is certainly a challenge to directly quantify energy expenditure or determine the presence of unbalanced ion concentrations in the CNS in vivo through non-invasive techniques. Zhu et al., designed the 31P MRS in vivo technique for the detection of intracellular content in the CNS of high-energy phosphate compounds, such as ATP or phosphocreatine, whose concentrations and relation to Pi can be altered under pathological conditions [148]. For their part, various authors have carried out in vivo studies using the sodium magnetic resonance technique (23NaMRI) to estimate pathological levels of sodium present in brain lesions of MS, and to determine the course of the disease in patients with the three clinical subtypes of MS (RRMS, SPMS, and PPMS) [149,150,151,152,153]. These studies highlight the importance of these novel MRI techniques in the study of MS and as potential biomarkers of neuroinflammation and degeneration [153].

4.4.4. Biomarkers of Glial Activation/Dysfunction

  • Nitric Oxide (NO)
NO is a signaling molecule in many physiological processes and in pathological processes such as MS. NO levels are increased in the CSF and serum of MS patients compared to individuals with non-inflammatory neurological disorders [159,160]. NO acts as a suppressor of mitochondrial activity by inhibiting cytochrome C oxidase, which decreases mitochondrial energy production [161]. NO has extremely toxic effects on neurons and glial cells, enhancing their apoptosis, and also affects the integrity and permeability of the BBB. This enables pro-inflammatory cells to reach the CNS. NO (X) degradation products, present in MS patients, are especially involved in the destruction of mitochondria, causing tissue hypoxia and accentuating damage in MS lesions [162].
  • Reactive Oxygen Species (ROS)
ROS are highly reactive oxygen species due to the presence of unpaired valence electrons. ROS alter the myelin sheaths and myelin-producing oligodendrocytes through oxidation mediated by their free radicals. Significant ROS generation was found in MS patients compared to healthy individuals, leading to cellular and tissue oxidative stress in their CNS [163]. ROS production in MS patients favors the generation of superoxide and peroxynitrite, which are harmful to glial and neuronal cells, accentuating the damage to the CNS. Likewise, higher levels of 7-ketocholesterol, a product of the oxidative degradation of lipids, were observed in the CSF of patients with MS, which in turn is capable of inducing neuronal damage through the activation and migration of microglial cells in CNS [164].
  • Glial fibrillary acidic protein (GFAP)
It has been observed that the expression of the intermediate filament protein of mature astrocytes GFAP increases in MS plaques, which would be expected to relate to a high level of astrocyte damage [165,166]. A higher level of GFAP was measured in CSF of patients with SPMS than in patients with RRMS, indicating greater neurological dysfunction in this type of MS, as well as a greater progression of disability [167]. Furthermore, MS patients with greater disabilities have higher GFAP levels compared to patients with lesser disabilities [124]. Even higher levels of GFAP are found during the MS relapse itself, and these levels are maintained for at least 5 weeks [168]. On the other hand, it has been observed that, in the CSF of patients with relapsed NMO, the level of GFAP is significantly higher than in the CSF of patients with relapsed MS [169,170]. This evidence suggests that GFAP can be used as a biomarker to detect the development of MS, indicating a high rate of astrogliosis, and could be used to differentiate between types of MS.
  • S100 calcium-binding protein B (S-100B)
S-100b protein is found in astroglia, a small subset of oligodendroglia, and a certain subgroup of neurons. The main function of this protein is to promote neuronal proliferation, oligodendrocyte differentiation, stimulation of calcium fluxes, maintaining astrocyte morphology, and facilitating the astrocyte and microglial activation that occurs intracellularly. Petzold et al. showed that an increasing amount of S100B levels were found in all MS subgroups which indicate cerebral injury [168].

5. Conclusions

The dearth of knowledge about the pathophysiology of MS and the clinical variation in MS subtypes makes it implausible to establish a single biomarker which guarantees the full evaluation of the disease. For a long time, numerous investigations have been conducted to identify potential biomarkers that provide meaningful information related to the development of the disease and the possible response of MS patients to treatments. Although progress has been made in the search for this ‘magical unique biomarker’, MS continues to be extremely unpredictable, with more unanswered questions than absolute certainties, the finding of it seems still a long way off. Therefore, to date, there remains no single reliable biomarker that can provide information on the prognosis of MS, distinguish between the different clinical courses of MS, and also predict responses to a specific form of treatment. Despite the various clinical challenges that a biomarker faces, continued advancement in research has resulted in the identification of a group of biomarkers specifically targeting the early stages of the disease. However, there remains a need for a panel of validated biomarkers that are capable of predicting and monitoring the efficiency of the growing number of treatment strategies available, with the aim of reducing the recurrence of relapses, and stopping the progression and disability of patients with MS.

Author Contributions

D.M. contributed by drafting and editing the manuscript; B.K.M. contributed by editing the manuscript; G.L.-R. contributed in review and editing the manuscript; S.R. contributed by drafting the manuscript; F.J.L.-I. and R.K. contributed by editing the manuscript; J.V. contributed by giving technical advice required for this manuscript; B.C. contributed in the conceptualization, supervision and editing the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work has been supported by a grant from the Health Institute Carlos III (PI20/01446) and FEDER funding.

Acknowledgments

The authors do not have any conflict of interest. R.K. acknowledged DBT for awarding Ramalingaswami Re-entry fellowship.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

APPAmyloid-Precursor Protein
AQP4Aquaporin-4 Protein
BBBBlood-Brain Barrier
CDMSClinically Defined MS
CHI3L1Chitinase-3-Like-1 Precursor
CISClinically Isolated Syndrome
CSFCerebro Spinal Fluid
DMTDisease-Modifying Therapies
DTIDiffusion Tensor Imaging
DWIDiffusion Weighted Imaging
EBNAEpstein-Barr Nuclear Antigen
FLAIRFLuid Attenuated Inversion Recovery Sequence
KFLCKappa Free Light Chain
LFLCLambda Free Light Chain
MBPMyelin Basic Protein
MOGMyelin Oligodendrocyte Glycoprotein
MSMultiple Sclerosis
MRIMagnetic Resonance Imaging
MRSMagnetic Resonance Spectroscopy
MTIMagnetization Transfer Imaging
MTRMagnetization Transfer Ratio
NAAN-Acetyl-Aspartate
NAWMNormal-Appearing White Matter
NFNeurofilaments
GFAPGlial Fibrillary Acidic Protein
NMOSDNeuromyelitis Optica Spectrum Disorders
OCBOligoclonal Bands
OCGBIgG-Type Oligoclonal Bands
OCMBIgM-Type Oligoclonal Bands
OCTOptical Coherence Tomography
PD-SEProton Density-Weighted Spin-Echo Imaging
PETPositron Emission Tomography
PPMSPrimary Progressive MS
RISRadiologically Isolated Syndrome
RNFLRetinal Nerve Fiber Layer
RRMSRelapsing Remitting MS
SPMSSecondary Progressive MS
S-100BS100 Calcium-Binding Protein B
VCAViral Capsid Antigen

References

  1. Dilokthornsakul, P.; Valuck, R.J.; Nair, K.V.; Corboy, J.R.; Allen, R.R.; Campbell, J.D. Multiple Sclerosis prevalence in the United States Commercially insured population. Neurology 2016, 86, 1014–1021. [Google Scholar] [CrossRef] [Green Version]
  2. Bhatia, R.; Bali, P.; Chowdhary, R. Epidemiology and genetic aspects of multiple sclerosis in India. Ann. Indian Acad. Neurol. 2015, 18, 6. [Google Scholar] [CrossRef]
  3. Huang, W.J.; Chen, W.W.; Zhang, X. Multiple sclerosis: Pathology, diagnosis and treatments. Exp. Ther. Med. 2017, 13, 3163–3166. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Lublin, F.D.; Reingold, S.C.; Cohen, J.A.; Cutter, G.R.; Sørensen, P.S.; Thompson, A.J.; Wolinsky, J.S.; Balcer, L.J.; Banwell, B.; Barkhof, F.; et al. Defining the clinical course of multiple sclerosis: The 2013 revisions. Neurology 2014, 83, 278–286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Weinshenker, B.G.; Bass, B.; Rice, G.P.; Noseworthy, J.; Carriere, W.; Baskerville, J.; Ebers, G.C. The natural history of multiple sclerosis: A geographically based study. I. Clinical course and disability. Brain 1989, 112, 133–146. [Google Scholar] [CrossRef] [PubMed]
  6. Confavreux, C.; Vukusic, S. Natural history of multiple sclerosis: A unifying concept. Brain 2006, 129, 606–616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Wallin, M.T.; Wilken, J.A.; Turner, A.P.; Williams, R.M.; Kane, R. Depression and multiple sclerosis: Review of a lethal combination. J. Rehabil. Res. Dev. 2006, 43, 45–46. [Google Scholar] [CrossRef] [PubMed]
  8. Gilgun-Sherki, Y.; Melamed, E.; Offen, D. The role of oxidative stress in the pathogenesis of multiple sclerosis: The need for effective antioxidant therapy. J. Neurol. 2004, 251, 261–268. [Google Scholar] [PubMed]
  9. Kleinschnitz, C.; Meuth, S.G.; Wiendl, H. The trials and errors in MS therapy. Int. MS J. 2008, 15, 79–80. [Google Scholar]
  10. Brilot, F.; Dale, R.C.; Selter, R.C.; Grummel, V.; Kalluri, S.R.; Aslam, M.; Selter, C.; Busch, V.; Zhou, D.; Cepok, S.; et al. Antibodies to native myelin oligodendrocyte glycoprotein in children with inflammatory demyelinating central nervous system disease. Ann. Neurol. 2009, 66, 833–842. [Google Scholar] [CrossRef]
  11. Lalive, P.H.; Häusler, M.G.; Maurey, H.; Mikaeloff, Y.; Tardieu, M.; Wiendl, H.; Schroeter, M.; Hartung, H.P.; Kieseier, B.C.; Menge, T. Highly reactive anti-myelin oligodendrocyte glycoprotein antibodies differentiate demyelinating diseases from viral encephalitis in children. Mult. Scler. 2011, 17, 297–302. [Google Scholar] [CrossRef] [PubMed]
  12. Baumann, M.; Sahin, K.; Lechner, C.; Hennes, E.M.; Schanda, K.; Mader, S.; Karenfort, M.; Selch, C.; Häusler, M.; Eisenkölbl, A.; et al. Clinical and neuroradiological differences of paediatric acute disseminating encephalomyelitis with and without antibodies to the myelin oligodendrocyte glycoprotein. J. Neurol. Neurosurg. Psychiatry 2015, 86, 265–272. [Google Scholar] [CrossRef] [PubMed]
  13. Ramanathan, S.; Dale, R.C.; Brilot, F. Anti-MOG antibody: The history, clinical phenotype, and pathogenicity of a serum biomarker for demyelination. Autoimmun. Rev. 2016, 15, 307–324. [Google Scholar] [CrossRef] [PubMed]
  14. Reindl, M.; Di Pauli, F.; Rostasy, K.; Berger, T. The spectrum of MOG autoantibody-associated demyelinating diseases. Nat. Rev. Neurol. 2013, 9, 455–461. [Google Scholar] [CrossRef] [PubMed]
  15. Paul, A.; Comabella, M.; Gandhi, R. Biomarkers in Multiple Sclerosis. Cold Spring Harb. Perspect. Med. 2019, 9, a029058. [Google Scholar] [CrossRef]
  16. Gul, M.; Jafari, A.A.; Shah, M.; Mirmoeeni, S.M.S.; Haider, S.U.; Moinuddin, S.; Chaudhry, A. Molecular Biomarkers in Multiple Sclerosis and Its Related Disorders: A Critical Review. Int. J. Mol. Sci. 2020, 21, 6020. [Google Scholar] [CrossRef]
  17. Porter, L.; Shoushtarizadeh, A.; Jelinek, G.A.; Brown, C.R.; Lim, C.K.; de Livera, A.M.; Jacobs, K.R.; Weiland, T.J. Metabolomic Biomarkers of Multiple Sclerosis: A Systematic Review. Front. Mol. Biosci. 2020, 7, 574133. [Google Scholar] [CrossRef]
  18. Toscano, S.; Patti, F. CSF biomarkers in multiple sclerosis: Beyond neuroinflammation. Neuroimmunol. Neuroinflamm. 2021, 8, 14–41. [Google Scholar]
  19. Jones, J.L.; Phuah, C.-L.; Cox, A.L.; Thompson, S.A.; Ban, M.; Shawcross, J.; Walton, A.; Sawcer, S.J.; Compston, A.; Coles, A.J. IL-21 drives secondary autoimmunity in patients with multiple sclerosis, following therapeutic lymphocyte depletion with alemtuzumab (Campath-1H). J. Clin. Investig. 2009, 119, 2052–2061. [Google Scholar] [CrossRef] [Green Version]
  20. Azzopardi, L.; Thompson, S.A.; Harding, K.E.; Cossburn, M.; Robertson, N.; Compston, A.; Coles, A.J.; Jones, J.L. Predicting autoimmunity after alemtuzumab treatment of multiple sclerosis. J. Neurol. Neurosurg. Psychiatry. 2013, 85, 795–798. [Google Scholar] [CrossRef] [Green Version]
  21. Berger, T.; Rubner, P.; Schautzer, F.; Egg, R.; Ulmer, H.; Mayringer, I.; Dilitz, E.; Deisenhammer, F.; Reindl, M. Antimyelin antibodies as a predictor of clinically definite multiple sclerosis after a first demyelinating event. N. Engl. J. Med. 2003, 349, 139–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Brickshawana, A.; Hinson, S.R.; Romero, M.F.; Lucchinetti, C.F.; Guo, Y.; Buttmann, M.; McKeon, A.; Pittock, S.J.; Chang, M.-H.; Chen, A.-P.; et al. Investigation of the KIR4.1 potassium channel as a putative antigen in patients with multiple sclerosis: A comparative study. Lancet Neurol. 2014, 13, 795–806. [Google Scholar] [CrossRef] [Green Version]
  23. Kuhle, J.; Pohl, C.; Mehling, M.; Edan, G.; Freedman, M.S.; Hartung, H.P.; Polman, C.H.; Miller, D.H.; Montalban, X.; Barkhof, F.; et al. Lack of association between antimyelin antibodies and progression to multiple sclerosis. N. Engl. J. Med. 2007, 356, 371–378. [Google Scholar] [CrossRef]
  24. Vastava, R.; Aslam, M.; Kalluri, S.; Schirmer, L.; Buck, D.; Tackenberg, B.; Rothhammer, V.; Chan, A.; Gold, R.; Berthele, A.; et al. Potassium channel KIR4.1 as an immune target in multiple sclerosis. N. Engl. J. Med. 2012, 367, 115–123. [Google Scholar]
  25. Corvol, J.C.; Pelletier, D.; Henry, R.G.; Caillier, S.J.; Wang, J.; Pappas, D.; Casazza, S.; Okuda, D.T.; Hauser, S.L.; Oksenberg, J.R.; et al. Abrogation of T cell quiescence characterizes patients at high risk for multiple sclerosis after the initial neurological event. Proc. Natl. Acad. Sci. USA 2008, 105, 11839–11844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Hedström, A.K.; Huang, J.; Michel, A.; Butt, J.; Brenner, N.; Hillert, J.; Waterboer, T.; Kockum, I.; Olsson, T.; Alfredsson, L. High Levels of Epstein–Barr virus nuclear antigen1-specific antibodies and infectious mononucleosis act both independently and synergistically to Increase multiple sclerosis risk. Front. Neurol. 2020, 10, 1368. [Google Scholar] [CrossRef] [PubMed]
  27. Abrahamyan, S.; Eberspächer, B.; Muna, H.M.M.; Hoshi, M.; Aly, L.; Luessi, F.; Groppa, S.; Klotzl, L.; Meuth, S.G.; Schroeder, C.; et al. Complete Epstein-Barr virus seropositivity in a large cohort of patients with early multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2020, 91, 681–686. [Google Scholar] [CrossRef]
  28. Dobson, R.; Kuhle, J.; Middeldorp, J.; Giovannoni, G. Epstein-Barr-negative MS: A true phenomenon? Neurol. Neuroimmunol. Neuroinflamm. 2017, 4, e318. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Zivadinov, R.; Bergsland, N.; Dwyer, M.G. Atrophied brain lesion volume, a magnetic resonance imaging biomarker for monitoring neurodegenerative changes in multiple sclerosis. Quant. Imaging Med. Surg. 2018, 8, 979–983. [Google Scholar] [CrossRef]
  30. Brex, P.A.; Parker, G.J. Lesion heterogeneity in multiple sclerosis: A study of the relations between appearances on T1 weighted images, T1 relaxation times and metabolite concentrations. J. Neurol. Neurosurg. Psychiatry 2000, 68, 627–632. [Google Scholar] [CrossRef] [Green Version]
  31. Sahraian, M.A.; Radue, E.W.; Haller, S.; Kappos, L. Black holes in multiple sclerosis: Definition, evolution, and clinical correlations. Acta Neurol. Scand. 2010, 122, 1–8. [Google Scholar] [CrossRef] [PubMed]
  32. Grossman, R.I.; Gonzalez-Scarano, F.; Atlas, S.W.; Galetta, S.; Silberberg, D.H. Multiple sclerosis: Gadolinium enhancement in MR imaging. Radiology 1986, 161, 721–725. [Google Scholar] [CrossRef]
  33. Kermode, A.G.; Thompson, A.J.; Tofts, P.; MacManus, D.G.; Kendall, B.E.; Kingsley, D.P.; Moseley, I.F.; Rudge, P.; McDonald, W.I. Breakdown of the blood-brain barrier precedes symptoms and other MRI signs of new lesions in multiple sclerosis. Pathogenetic and clinical implications. Brain 1990, 113, 1477–1489. [Google Scholar] [CrossRef]
  34. Schmidt, C.; Hattingen, E.; Faehndrich, J.; Jurcoane, A.; Porto, L. Detectability of multiple sclerosis lesions with 3T MRI: A comparison of proton-density weighted and FLAIR sequence. J. Neuroradiol. 2012, 39, 52–57. [Google Scholar] [CrossRef] [PubMed]
  35. Trip, S.A.; Miller, D.H. Imaging in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 2005, 76, iii11–iii18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Chong, A.L.; Chandra, R.V.; Chuah, K.C.; Roberts, E.L.; Stuckey, S.L. Proton density MRI increases detection of cervical spinal cord multiple sclerosis lesions compared with T2-weighted fast spin-echo. AJNR Am. J. Neuroradiol. 2016, 37, 180–184. [Google Scholar] [CrossRef] [Green Version]
  37. Moccia, M.; Van de Pavert, S.; Eshaghi, A.; Haider, L.; Pichat, J.; Yiannakas, M.; Ourselin, S.; Wang, Y.; Wheeler-Kingshott, C.; Thompson, A.; et al. Pathologic correlates of the magnetization transfer ratio in multiple sclerosis. Neurology 2020, 95, e2965–e2976. [Google Scholar] [CrossRef]
  38. Dousset, V.; Grossman, R.I.; Ramer, K.N.; Schnall, M.D.; Young, L.H.; Gonzalez-Scarano, F.; Lavi, E.; Cohen, J.A. Experimental allergic encephalomyelitis and multiple sclerosis: Lesion characterization with magnetization transfer imaging. Radiology 1992, 182, 483–491. [Google Scholar] [CrossRef]
  39. Deloire-Grassin, M.S.; Brochet, B.; Quesson, B.; Delalande, C.; Dousset, V.; Canioni, P.; Petry, K.G. In vivo evaluation of remyelination in rat brain by magnetization transfer imaging. J. Neurol. Sci. 2000, 178, 10–16. [Google Scholar] [CrossRef]
  40. Trip, S.A.; Schlottmann, P.G.; Jones, S.J.; Li, W.Y.; Garway-Heath, D.F.; Thompson, A.J.; Plant, G.T.; Miller, D.H. Optic nerve magnetization transfer imaging and measures of axonal loss and demyelination in optic neuritis. Mult. Scler. 2007, 13, 875–879. [Google Scholar] [CrossRef]
  41. Avila, M.G.S.; Claudio, A.O.; Zabala, E.L.; Teledo, J.D. Diffusion weighted imaging changes in multiple sclerosis patients, frequency and co-relation to disease activity. Austin Neurol. 2018, 3, 1012. [Google Scholar]
  42. Abolhasani Foroughi, A.; Salahi, R.; Nikseresht, A.; Heidari, H.; Nazeri, M.; Khorsand, A. Comparison of diffusion-weighted imaging and enhanced T1-weighted sequencing in patients with multiple sclerosis. Neuroradiol. J. 2017, 30, 347–351. [Google Scholar] [CrossRef] [PubMed]
  43. Aung, W.Y.; Mar, S.; Benzinger, T.L. Diffusion tensor MRI as a biomarker in axonal and myelin damage. Imaging Med. 2013, 5, 427–440. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Pierpaoli, C.; Jezzard, P.; Basser, P.J.; Barnett, A.; Di Chiro, G. Diffusion tensor MR imaging of the human brain. Radiology 1996, 201, 637–648. [Google Scholar] [CrossRef] [PubMed]
  45. Liu, Y.; Mitchell, P.J.; Kilpatrick, T.J.; Stein, M.S.; Harrison, L.C.; Baker, J.; Ditchfield, M.; Li, K.; Egan, G.F.; Butzkueven, H.; et al. Diffusion tensor imaging of acute inflammatory lesion evolution in multiple sclerosis. J. Clin. Neurosci. 2012, 19, 1689–1694. [Google Scholar] [CrossRef] [PubMed]
  46. Tian, W.; Zhu, T.; Zhong, J.; Liu, X.; Rao, P.; Segal, B.M.; Ekholm, S. Progressive decline in fractional anisotropy on serial DTI examinations of the corpus callosum: A putative marker of disease activity and progression in SPMS. Neuroradiology 2012, 54, 287–297. [Google Scholar] [CrossRef]
  47. Hasan, K.M.; Gupta, R.K.; Santos, R.M.; Wolinsky, J.S.; Narayana, P.A. Diffusion tensor fractional anisotropy of the normal-appearing seven segments of the corpus callosum in healthy adults and relapsing-remitting multiple sclerosis patients. J. Magn. Reson. Imaging 2005, 21, 735–743. [Google Scholar] [CrossRef]
  48. Hakulinen, U.; Brander, A.; Ryymin, P.; Öhman, J.; Soimakallio, S.; Helminen, M.; Dastidar, P.; Eskola, H. Repeatability and variation of region-of-interest methods using quantitative diffusion tensor MR imaging of the brain. BMC Med. Imaging 2012, 12, 30. [Google Scholar] [CrossRef] [Green Version]
  49. Akbar, N.; Rudko, D.A.; Parmar, K. Magnetic resonance imaging of multiple sclerosis. Sci. J. Mult. Scler. 2017, 1, 008–020. [Google Scholar]
  50. Narayana, P.A. Magnetic resonance spectroscopy in the monitoring of multiple sclerosis. J. Neuroimaging. 2005, 15, 46S–57S. [Google Scholar] [CrossRef]
  51. Grazioli, E.; Zivadinov, R.; Weinstock-Guttman, B.; Lincoff, N.; Baier, M.; Wong, J.R.; Hussein, S.; Cox, J.L.; Hojnacki, D.; Ramanathan, M. Retinal nerve fiber layer thickness is associated with brain MRI outcomes in multiple sclerosis. J. Neurol. Sci. 2008, 268, 12–17. [Google Scholar] [CrossRef]
  52. Britze, J.; Frederiksen, J.L. Optical coherence tomography in multiple sclerosis. Eye 2018, 32, 884–888. [Google Scholar] [CrossRef] [Green Version]
  53. Oh, U.; Fujita, M.; Ikonomidou, V.N.; Evangelou, I.E.; Matsuura, E.; Harberts, E.; Fujimura, Y.; Richert, N.D.; Ohayon, J.; Pike, V.W.; et al. Translocator protein PET imaging for glial activation in multiple sclerosis. J. Neuroimm. Pharmacol. 2011, 6, 354–361. [Google Scholar] [CrossRef] [Green Version]
  54. Stankoff, B.; Poirion, E.; Tonietto, M.; Bodini, B. Exploring the heterogeneity of MS lesions using positron emission tomography: A reappraisal of their contribution to disability. Brain Path. 2018, 28, 723–734. [Google Scholar] [CrossRef]
  55. Hinsinger, G.; Galéotti, N.; Nabholz, N.; Urbach, S.; Rigau, V.; Demattei, C.; Lehmann, S.; Camu, W.; Labauge, P.; Castelnovo, G.; et al. Chitinase 3-like proteins as diagnostic and prognostic biomarkers of multiple sclerosis. Mult. Scler. 2015, 21, 1251–1261. [Google Scholar] [CrossRef] [PubMed]
  56. Cantó, E.; Reverter, F.; Morcillo-Suárez, C.; Matesanz, F.; Fernández, O.; Izquierdo, G.; Vandenbroeck, K.; Rodríguez-Antigüedad, A.; Urcelay, E.; Arroyo, R.; et al. Chitinase 3-like 1 plasma levels are increased in patients with progressive forms of multiple sclerosis. Mult. Scler. 2012, 8, 983–990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Burman, J.; Raininko, R.; Blennow, K.; Zetterberg, H.; Axelsson, M.; Malmestrom, C. YKL-40 is a CSF biomarker of intrathecal inflammation in secondary progressive multiple sclerosis. J. Neuroimmunol. 2016, 292, 52–57. [Google Scholar] [CrossRef] [Green Version]
  58. Khademi, M.; Kockum, I.; Andersso, M.L.; Iacobaeus, E.; Brundin, L.; Sellebjerg, F.; Hillert, J.; Piehl, F.; Olsson, T. Cerebrospinal Fluid CXCL13 in multiple sclerosis: A suggestive prognostic marker for the disease course. Mult. Scler. J. 2010, 17, 335–343. [Google Scholar] [CrossRef] [PubMed]
  59. Mouzaki, A.; Rodi, M.; Dimisianos, N.; Emmanuil, A.; Kalavrizioti, D.; Lagoudaki, R.; Grigoriadis, N.C.; Papathanasopoulos, P. Immune Parameters That Distinguish Multiple Sclerosis Patients from Patients with Other Neurological Disorders at Presentation. PLoS ONE 2015, 10, e0135434. [Google Scholar]
  60. Kim, B.S.; Jin, Y.H. IL-1 signal affects both protection and pathogenesis of virus-induced chronic CNS demyelinating disease. J. Neuroinflammation 2012, 9, 217. [Google Scholar]
  61. Huang, J.; Khademi, M.; Fugger, L.; Lindhe, Ö.; Novakova, L.; Axelsson, M.; Malmeström, C.; Constantinescu, C.; Lycke, J.; Piehl, F.; et al. Inflammation-related plasma and CSF biomarkers for multiple sclerosis. Proc. Natl. Acad. Sci. USA 2020, 117, 12952–12960. [Google Scholar] [CrossRef]
  62. Bai, Z.; Chen, D. Cerebrospinal fluid and blood cytokines as biomarkers for multiple sclerosis: A systematic review and meta-analysis of 226 studies with 13,526 patients. Front. Neurosci. 2019, 13, 1026. [Google Scholar] [CrossRef]
  63. Chen, Y.C.; Yang, X.; Miao, L. Serum level of interleukin-6 in Chinese patients with multiple sclerosis. J. Immunol. 2012, 249, 109–111. [Google Scholar] [CrossRef] [PubMed]
  64. Ozenci, V.; Kouwenhoven, M.; Huang, Y.M.; Kivisakk, P.; Link, H. Multiple sclerosis is associated with an imbalance between tumor necrosis factor -alpha and IL- 10 secreting blood cells that is corrected by interferon-beta treatment. Clin. Exp. Immunol. 2000, 120, 147–153. [Google Scholar] [CrossRef]
  65. Ascherio, A.; Munger, K.L. Environmental risk factors for multiple sclerosis. Part I: The role of infection. Ann. Neurol. 2007, 61, 288–299. [Google Scholar] [CrossRef] [PubMed]
  66. Leibovitch, E.C.; Jacobson, S. Evidence linking HHV-6 with multiple sclerosis: An update. Curr. Opin. Virol. 2014, 9, 127–133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Merelli, E.; Bedin, R.; Sola, P.; Barozzi, P.; Mancardi, G.L.; Ficarra, G.; Franchini, G. Human herpes virus 6 and human herpes virus 8 DNA sequences in brains of multiple sclerosis patients, normal adults and children. J. Neurol. 1997, 244, 450–454. [Google Scholar] [CrossRef]
  68. Barres, B.A.; Schmid, R.; Sendnter, M.; Raff, M.C. Multiple extracellular signals are required for long-term oligodendrocyte survival. Development 1993, 118, 283–295. [Google Scholar] [CrossRef]
  69. Opsahl, M.L.; Kennedy, P.G. Early and late HHV-6 gene transcripts in multiple sclerosis lesions and normal appearing white matter. Brain 2005, 128, 516–527. [Google Scholar] [CrossRef] [Green Version]
  70. Dominguez-Mozo, M.I.; Perez-Perez, S.; Villar, L.M.; Oliver-Martos, B.; Villarrubia, N.; Matesanz, F.; Costa-Frossard, L.; Pinto-Medel, M.J.; García-Sánchez, M.I.; Ortega-Madueño, I.; et al. Predictive factors and early biomarkers of response in multiple sclerosis patients treated with natalizumab. Sci. Rep. 2020, 10, 14244. [Google Scholar] [CrossRef]
  71. Pryce, G.; Baker, D. Oligoclonal bands in multiple sclerosis; Functional significance and therapeutic implications. Does the specificity matter? Mult. Scler. Relat. Disord. 2018, 25, 131–137. [Google Scholar] [CrossRef]
  72. Trbojevic-Cepe, M. Detection of oligoclonal Ig bands: Clinical significance and trends in methodological improvement. EJIFCC 2004, 15, 86–94. [Google Scholar]
  73. Ziemssen, T.; Ziemssen, F. The role of the humoral immune system in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Autoimmun. Rev. 2005, 4, 460–467. [Google Scholar] [CrossRef]
  74. Kuhle, J.; Disanto, G.; Dobson, R.; Adiutori, R.; Bianchi, L.; Topping, J.; Bestwick, J.P.; Meier, U.-C.; Marta, M.; Dalla Costa, G.; et al. Conversion from clinically isolated syndrome to multiple sclerosis: A large multicentre study. Mult. Scler. 2015, 21, 1013–1024. [Google Scholar] [CrossRef]
  75. Matute-Blanch, C.; Villar, L.M.; Álvarez-Cermeño, J.C.; Rejdak, K.; Evdoshenko, E.; Makshakov, G.; Nazarov, V.; Lapin, S.; Midaglia, L.; Vidal-Jordana, A.; et al. Neurofilament light chain and oligoclonal bands are prognostic biomarkers in radiologically isolated syndrome. Brain 2018, 141, 1085–1093. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Dobson, R.; Ramagopalan, S.; Davis, A.; Giovannoni, G. Cerebrospinal fluid oligoclonal bands in multiple sclerosis and clinically isolated syndromes: A meta-analysis of prevalence, prognosis and effect of latitude. J. Neurol. Neurosurg. Psychiatry 2013, 84, 909–914. [Google Scholar] [CrossRef] [PubMed]
  77. Katsavos, S.; Anagnostouli, M. Biomarkers in Multiple Sclerosis: An up-to-date overview. Mult. Scler. Int. 2013, 2013, 340508. [Google Scholar] [CrossRef] [Green Version]
  78. Nilsson, P.; Larsson, E.M.; Maly-Sundgren, P.; Perfekt, R.; Sandberg-Wollheim, M. Predicting the outcome of optic neuritis: Evaluation of risk factors after 30 years of follow-up. J. Neurol. 2005, 252, 396–402. [Google Scholar] [CrossRef] [PubMed]
  79. Tintore, M.; Rovira, À.; Río, J.; Otero-Romero, S.; Arrambide, G.; Tur, C.; Comabella, M.; Nos, C.; Arévalo, M.J.; Negrotto, L.; et al. Defining high, medium and low impact prognostic factors for developing multiple sclerosis. Brain 2015, 138, 1863–1874. [Google Scholar] [CrossRef] [Green Version]
  80. Skov, A.G.; Skov, T.; Frederiksen, J.L. Oligoclonal bands predict multiple sclerosis after optic neuritis: A literature survey. Mult. Scler. 2011, 17, 404–410. [Google Scholar] [CrossRef]
  81. Söderström, M.; Ya-Ping, J.; Hillert, J.; Link, H. Optic neuritis: Prognosis for multiple sclerosis from MRI, CSF, and HLA findings. Neurology 1998, 50, 708–714. [Google Scholar] [CrossRef]
  82. Freedman, M.S.; Thompson, E.J.; Deisenhammer, F.; Giovannoni, G.; Grimsley, G.; Keir, G.; Ohman, S.; Racke, M.K.; Sharief, M.; Sindic, C.J.; et al. Recommended standard of cerebrospinal fluid analysis in the diagnosis of multiple sclerosis: A consensus statement. Arch. Neurol. 2005, 62, 865–870. [Google Scholar] [CrossRef] [Green Version]
  83. Farina, G.; Magliozzi, R.; Pitteri, M.; Reynolds, R.; Rossi, S.; Gajofatto, A.; Benedetti, M.D.; Facchiano, F.; Monaco, S.; Calabrese, M. Increased cortical lesion load and intrathecal inflammation is associated with oligoclonal bands in multiple sclerosis patients: A combined CSF and MRI study. J. Neuroinflammation 2017, 14, 40. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Graner, M.; Pointon, T.; Manton, S.; Green, M.; Dennison, K.; Davis, M.; Braiotta, G.; Craft, J.; Edwards, T.; Polonsky, B.; et al. Oligoclonal IgG antibodies in multiple sclerosis target patient-specific peptides. PLoS ONE 2020, 15, e0228883. [Google Scholar]
  85. Rojas, J.I.; Patrucco, L.; Tizio, S.; Cristiano, E. Oligoclonal bands in the cerebrospinal fluid and increased brain atrophy in early stages of relapsing-remitting multiple sclerosis. Arq. Neuropsiquiatr. 2012, 70, 574–577. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ferreira, D.; Voevodskaya, O.; Imrell, K.; Stawiarz, L.; Spulber, G.; Wahlund, L.O.; Hillert, J.; Westman, E.; Karrenbauer, V.D. Multiple sclerosis patients lacking oligoclonal bands in the cerebrospinal fluid have less global and regional brain atrophy. J. Neuroinmun. 2014, 274, 149–154. [Google Scholar] [CrossRef] [PubMed]
  87. Villar, L.M.; Sádaba, M.C.; Roldán, E.; Masjuan, J.; González-Porqué, P.; Villarrubia, N.; Espiño, M.; García-Trujillo, J.A.; Bootello, A.; Alvarez-Cermeño, J.C. Intrathecal synthesis of oligoclonal IgM against myelin lipids predicts an aggressive disease course in MS. J. Clin. Investig. 2005, 115, 187–194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Thompson, A.J.; Banwell, B.L.; Barkhof, F.; Carroll, W.M.; Coetzee, T.; Comi, G.; Correale, J.; Fazekas, F.; Filippi, M.; Freedman, M.S.; et al. Diagnosis of multiple sclerosis: 2017 revisions of the McDonald criteria. Lancet Neurol. 2018, 17, 162–173. [Google Scholar] [CrossRef]
  89. Villar, L.; García-Barragán, N.; Espiño, M.; Roldán, E.; Sádaba, M.; Gómez-Rial, J.; González-Porqué, P.; Alvarez-Cermeño, J. Influence of oligoclonal IgM specificity in multiple sclerosis disease course. Mult. Scler. 2008, 14, 183–187. [Google Scholar] [CrossRef]
  90. Monreal, E.; Sainz de la Maza, S.; Costa-Frossard, L.; Walo-Delgado, P.; Zamora, J.; Fernández-Velasco, J.I.; Villarrubia, N.; Espiño, M.; Lourido, D.; Lapuente, P.; et al. Predicting aggressive multiple sclerosis with intrathecal IgM synthesis among patients with a clinically isolated syndrome. Neurol. Neuroimmunol. Neuroinflamm. 2021, 8, e1047. [Google Scholar] [CrossRef]
  91. Thangarajh, M.; Gomez-Rial, J.; Hedström, A.K.; Hillert, J.; Alvarez-Cermeño, J.C.; Masterman, T.; Villar, L.M. Lipid-specific immunoglobulin M in CSF predicts adverse long-term outcome in multiple sclerosis. Mult. Scler. 2008, 14, 1208–12013. [Google Scholar] [CrossRef]
  92. Álvarez-Cermeño, J.C.; Muñoz-Negrete, F.J.; Costa-Frossard, L.; Sainz de la Maza, S.; Villar, L.M.; Rebolleda, G. Intrathecal lipid-specific oligoclonal IgM synthesis associates with retinal axonal loss in multiple sclerosis. J. Neurol. Sci. 2016, 360, 41–44. [Google Scholar] [CrossRef]
  93. Villar, L.M.; Picón, C.; Costa-Frossard, L.; Alenda, R.; García-Caldentey, J.; Espiño, M.; Muriel, A.; Álvarez-Cermeño, J.C. Cerebrospinal fluid immunological biomarkers associated with axonal damage in multiple sclerosis. Eur. J. Neurol. 2015, 22, 1169–1175. [Google Scholar] [CrossRef] [PubMed]
  94. Villar, L.M.; Casanova, B.; Ouamara, N.; Comabella, M.; Jalili, F.; Leppert, D.; de Andrés, C.; Izquierdo, G.; Arroyo, R.; Avşar, T.; et al. Immunoglobulin M oligoclonal bands: Biomarker of targetable inflammation in primary progressive multiple sclerosis. Ann. Neurol. 2014, 76, 231–240. [Google Scholar] [CrossRef] [PubMed]
  95. Yuan, A.; Rao, M.V.; Nixon, R.A. Neurofilaments at a glance. J. Cell Sci. 2012, 125, 3257–3263. [Google Scholar] [CrossRef] [PubMed]
  96. Khalil, M.; Teunissen, C.E.; Otto, M.; Pieh, L.F.; Sormani, M.P.; Gattringer, T.; Barro, C.; Kappos, L.; Comabella, M.; Fazekas, F.; et al. Neurofilaments as biomarkers in neurological disorders. Nature Rev. Neurol. 2018, 14, 577–589. [Google Scholar] [CrossRef] [PubMed]
  97. Salzer, J.; Svenningsson, A.; Sundstrom, P. Neurofilament light as a prognostic marker in multiple sclerosis. Mult. Scler. 2010, 16, 287–292. [Google Scholar] [CrossRef] [PubMed]
  98. Arrambide, G.; Espejo, C.; Eixarch, H. Neurofilament light chain level is a weak risk factor for the development of MS. Neurology 2016, 87, 1076–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Modvig, S.; Degn, M.; Roed, H.; Sørensen, T.L.; Larsson, H.B.; Langkilde, A.R.; Frederiksen, J.L.; Sellebjerg, F. Cerebrospinal fluid levels of chitinase 3-like 1 and neurofilament light chain predict multiple sclerosis development and disability after optic neuritis. Mult. Scler. 2015, 21, 1761–1770. [Google Scholar] [CrossRef] [PubMed]
  100. Barro, C.; Leocani, L.; Leppert, D.; Comi, G.; Kappos, L.; Kuhle, J. Fluid biomarker and electrophysiological outcome measures for progressive MS trials. Mult. Scler. 2017, 23, 1600–1613. [Google Scholar] [CrossRef]
  101. Novakova, L.; Zetterberg, H.; Sundström, P.; Axelsson, M.; Khademi, M.; Gunnarsson, M.; Malmeström, C.; Svenningsson, A.; Olsson, T.; Piehl, F.; et al. Monitoring disease activity in multiple sclerosis using serum neurofilament light protein. Neurology 2017, 89, 2230–2237. [Google Scholar] [CrossRef] [Green Version]
  102. Gunnarsson, M.; Malmeström, C.; Axelsson, M.; Sundström, P.; Dahle, C.; Vrethem, M.; Olsson, T.; Piehl, F.; Norgren, N.; Rosengren, L.; et al. Axonal damage in relapsing multiple sclerosis is markedly reduced by natalizumab. Ann. Neurol. 2011, 69, 83–89. [Google Scholar] [CrossRef]
  103. Kuhle, J.; Kropshofer, H.; Haering, D.A.; Kundu, U.; Meinert, R.; Barro, C.; Dahlke, F.; Tomic, D.; Leppert, D.; Kappos, L. Blood neurofilament light chain as a biomarker of MS disease activity and treatment response. Neurology 2019, 92, e1007–e1015. [Google Scholar] [CrossRef]
  104. Petzold, A. Neurofilament phosphoforms: Surrogate markers for axonal injury, degeneration and loss. J. Neurol. Sci. 2005, 233, 183–198. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Teunissen, C.E.; Iacobaeus, E.; Khademi, M.; Brundin, L.; Norgren, N.; Koel-Simmelink, M.J.; Schepens, M.; Bouwman, F.; Twaalfhoven, H.A.; Blom, H.J.; et al. Combination of CSF N-acetylaspartate and neurofilaments in multiple sclerosis. Neurology 2009, 72, 1322–1329. [Google Scholar] [CrossRef] [PubMed]
  106. Cohen, S.R.; Herndon, R.M.; McKhann, G.M. Radioimmunoassay of myelin basic protein in spinal fluid: An index of active demyelination. N. Engl. J. Med. 1976, 295, 1455–1457. [Google Scholar] [CrossRef] [PubMed]
  107. Whitaker, J.N. Myelin encephalitogenic protein fragments in cerebrospinal fluid of persons with multiple sclerosis. Neurology 1977, 27, 911–920. [Google Scholar] [CrossRef]
  108. Romme Christensen, J.; Börnsen, L.; Khademi, M.; Olsson, T.; Jensen, P.E.; Sørensen, P.S. Sellebjerg F CSF inflammation and axonal damage are increased and correlate in progressive multiple sclerosis. Mult. Scler. 2013, 19, 877–884. [Google Scholar] [CrossRef] [Green Version]
  109. Harris, V.K.; Sadiq, S.A. Disease biomarkers in multiple sclerosis: Potential for use in therapeutic decision making. Mol. Diagn. Ther. 2009, 13, 225–244. [Google Scholar] [CrossRef]
  110. Wekerle, H.; Lassmann, H. The immunology of inflammatory demyelinating disease. In McAlpine’s Multiple Sclerosis, 4th ed.; Compston, A., Ed.; Elsevier Health Sciences: Amsterdam, The Netherlands, 2005. [Google Scholar]
  111. Sellebjerg, F.; Christiansen, M.; Garred, P. MBP, anti-MBP and anti-PLP antibodies, and intrathecal complement activation in multiple sclerosis. Mult. Scler. 1998, 4, 127–131. [Google Scholar] [CrossRef]
  112. O’Conner, K.C.; Lopez-Amaya, C.; Gagne, D.; Lovato, L.; Moore-Odom, N.H.; Kennedy, J.; Krupp, L.; Tenembaum, S.; Ness, J.; Belman, A.; et al. Antimyelin antibodies modulate clinical expression of childhood Multiple Sclerosis. J. Immunol. 2010, 223, 92–99. [Google Scholar]
  113. Spadaro, M.; Gerdes, L.A.; Krumbholz, M.; Ertl-Wagner, B.; Thaler, F.S.; Schuh, E.; Metz, I.; Blaschek, A.; Dick, A.; Bruck, W.; et al. Autoantibodies to MOG in a distinct subgroup of adult Multiple Sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2016, 3, e257. [Google Scholar] [CrossRef] [Green Version]
  114. Kuerten, S.; Lanz, T.V.; Lingampalli, N.; Lahey, L.J.; Kleinschnitz, C.; Mäurer, M.; Schroeter, M.; Braune, S.; Ziemssen, T.; Ho, P.P.; et al. Autoantibodies against central nervous system antigens in a subset of B cell-dominant multiple sclerosis patients. Proc. Natl. Acad. Sci. USA 2020, 117, 21512–21518. [Google Scholar] [CrossRef] [PubMed]
  115. Ziemssen, T.; Akgün, K.; Brück, W. Molecular biomarkers in multiple sclerosis. J. Neuroinflammation 2019, 16, 272. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Lefvert, A.K.; Link, H. IgG production within the central nervous system: A critical review of proposed formulae. Ann. Neurol. 1985, 17, 13–20. [Google Scholar] [CrossRef] [PubMed]
  117. LeVine, S.M. Albumin and multiple sclerosis. BMC Neurol. 2016, 16, 47. [Google Scholar] [CrossRef] [Green Version]
  118. Harris, V.K.; Tuddenham, J.F.; Sadiq, S.A. Biomarkers of multiple sclerosis: Current findings. Degener. Neurol. Neuromuscul. Dis. 2017, 7, 19–29. [Google Scholar] [CrossRef] [Green Version]
  119. Ahlbrecht, J.; Martino, F.; Pul, R.; Skritpuletz, T.; Suhs, K.W.; Schauerte, C.; Yildiz, O.; Trebst, O.; Tasto, L.; Thum, S.; et al. Deregulation of microRNA-181C in cerebrospinal fluid of patients clinically isolated syndrome is associated with early conversion to Relapsing-Remitting Multiple Sclerosis. Mult. Scler. 2016, 22, 1202–1214. [Google Scholar] [CrossRef]
  120. Bergman, P.; Piket, E.; Khademi, M.; James, T.; Brundin, L.; Olsson, T.; Piehl, F.; Jagodic, M. Circulating miR-150 in CSF is a novel candidate biomarker for multiple sclerosis. Neurol. Neuroimmunol. Neuroinflamm. 2016, 3, e219. [Google Scholar] [CrossRef] [Green Version]
  121. Ma, X.; Zhou, J.; Zhong, Y. Expression, regulation and functions of microRNAs in multiple sclerosis. Int. J. Med. Sci. 2014, 11, 810–818. [Google Scholar] [CrossRef] [Green Version]
  122. Gresle, M.M.; Shaw, G.; Jarrott, B.; Alexandrou, E.N.; Friedhuber, A.; Kilpatrick, T.J.; Butzkueven, H. Validation of a novel biomarker for acute axonal injury in experimental autoimmune encephalomyelitis. J. Neurosci. Res. 2008, 86, 3548–3555. [Google Scholar] [CrossRef] [PubMed]
  123. Petzold, A.; Eikelenboom, M.J.; Keir, G.; Grant, D.; Lazeron, R.H.; Polman, C.H.; Uitdehaag, B.M.; Thompson, E.J.; Giovannoni, G. Axonal damage accumulates in the progressive phase of multiple sclerosis: Three years follow up study. J. Neurol. Neurosurg. Psychiatry 2005, 76, 206–211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Brettschneider, J.; Petzold, A.; Junker, A.; Tumani, H. Axonal damage markers in the cerebrospinal fluid of patients with clinically isolated syndrome improve predicting conversion to definite multiple sclerosis. Mult. Scler. 2006, 12, 143–148. [Google Scholar] [CrossRef] [PubMed]
  125. Rejdak, K.; Petzold, A.; Stelmasiak, Z.; Giovannoni, G. Cerebrospinal fluid brain specific proteins in relation to nitric oxide metabolites during relapse of multiple sclerosis. Mult. Scler. 2008, 14, 59–66. [Google Scholar] [CrossRef]
  126. Lycke, J.N.; Karlsson, J.E.; Andersen, O.; Rosengren, L.E. Neurofilament protein in cerebrospinal fluid: A potential marker of activity in multiple sclerosis. J. Neurol. Neurosurg. Psychiatry 1998, 64, 402–404. [Google Scholar] [CrossRef] [Green Version]
  127. Malmeström, C.; Haghighi, S.; Rosengren, L.; Andersen, O.; Lycke, J. Neurofilament light protein and glial fibrillary acidic protein as biological markers in MS. Neurology 2003, 61, 1720–1725. [Google Scholar] [CrossRef] [PubMed]
  128. Teunissen, C.E.; Dijkstra, P.C.; Polman, C. Biological markers in CSF and blood for axonal degeneration in multiple sclerosis. Lancet Neurol. 2005, 4, 32–41. [Google Scholar] [CrossRef]
  129. Bandyopadhyay, B.; Li, G.; Yin, H.; Kuret, J. Tau aggregation and toxicity in a cell culture model of tauopathy. J. Biol. Chem. 2007, 282, 16454–16464. [Google Scholar] [CrossRef] [Green Version]
  130. Anderson, J.M.; Patani, R.; Reynolds, R.; Nicholas, R.; Compston, A.; Spillantini, M.G.; Chandran, S. Evidence for abnormal tau phosphorylation in early aggressive multiple sclerosis. Acta Neuropathol. 2009, 117, 583–589. [Google Scholar] [CrossRef]
  131. Sjögren, M.; Vanderstichele, H.; Agren, H.; Zachrisson, O.; Edsbagge, M.; Wikkelsø, C.; Skoog, I.; Wallin, A.; Wahlund, L.O.; Marcusson, J.; et al. Tau and Abeta42 in cerebrospinal fluid from healthy adults 21-93 years of age: Establishment of reference values. Clin. Chem. 2001, 47, 1776–1781. [Google Scholar] [CrossRef] [Green Version]
  132. Anderson, J.M.; Patani, R.; Reynolds, R.; Nicholas, R.; Compston, A.; Spillantini, M.G.; Chandran, S. Abnormal tau phosphorylation in primary progressive multiple sclerosis. Acta Neuropath. 2010, 119, 591–600. [Google Scholar] [CrossRef]
  133. Brettschneider, J.; Maier, M.; Arda, S.; Claus, A.; Süssmuth, S.D.; Kassubek, J.; Tumani, H. Tau protein level in cerebrospinal fluid is increased in patients with early multiple sclerosis. Mult. Scler. 2005, 11, 261–265. [Google Scholar] [CrossRef]
  134. Tiberio, M.; Chard, D.T.; Altmann, D.R.; Davies, G.; Griffin, C.M.; McLean, M.A.; Rashid, W.; Sastre-Garriga, J.; Thompson, A.J.; Miller, D.H. Metabolite changes in early relapsing-remitting multiple sclerosis: A two-year follow-up study. J. Neurol. 2006, 253, 224–230. [Google Scholar] [CrossRef] [PubMed]
  135. Van Au Duong, M.; Audoin, B.; Le Fur, Y.; Confort-Gouny, S.; Malikova, I.; Soulier, E.; Viout, P.; Ali-Cherif, A.; Pelletier, J.; Cozzone, P.J.; et al. Relationships between gray matter metabolic abnormalities and white matter inflammation in patients at the very early stage of MS: A MRSI study. J. Neurol. 2007, 254, 914–923. [Google Scholar] [CrossRef] [PubMed]
  136. Viala, K.; Stievenart, J.L.; Cabanis, E.A.; Lyon-Caen, O.; Tourbah, A. Study with localized proton magnetic resonance spectroscopy of 31 multiple sclerosis lesions: Correlations with clinical and MRI features. Rev. Neurol. 2001, 157, 35–44. [Google Scholar]
  137. Narayanan, S.; De Stefano, N.; Francis, G.S.; Arnaoutelis, R.; Caramanos, Z.; Collins, D.L.; Pelletier, D.; Arnason, B.G.W.; Antel, J.P.; Arnold, D.L. Axonal metabolic recovery in multiple sclerosis patients treated with interferon beta-1b. J. Neurol. 2001, 248, 979–986. [Google Scholar] [CrossRef] [PubMed]
  138. Povlishock, J.T. Traumatically induced axonal injury: Pathogenesis and pathobiological implications. Brain Pathol. 1992, 2, 1–12. [Google Scholar]
  139. Adams, R.D.; Kubik, C.S. The morbid anatomy of the demyelinative disease. Am. J. Med. 1952, 12, 510–546. [Google Scholar] [CrossRef]
  140. Mattsson, N.; Axelsson, M.; Haghighi, S.; Malmeström, C.; Wu, G.; Anckarsäter, R.; Sankaranarayanan, S.; Andreasson, U.; Fredrikson, S.; Gundersen, A.; et al. Reduced cerebrospinal fluid BACE1 activity in multiple sclerosis. Mult. Scler. 2009, 15, 448–454. [Google Scholar] [CrossRef]
  141. Gehrmann, J.; Banati, R.B.; Cuzner, M.L.; Kreutzberg, G.W.; Newcombe, J. Amyloid precursor protein (APP) expression in multiple sclerosis lesions. Glia 1995, 15, 141–151. [Google Scholar] [CrossRef]
  142. Clarner, T.; Buschmann, J.P.; Beyer, C.; Kipp, M. Glial amyloid precursor protein expression is restricted to astrocytes in an experimental toxic model of multiple sclerosis. J. Mol. Neurosci. 2011, 43, 268–274. [Google Scholar] [CrossRef]
  143. Young, E.A.; Fowler, C.D.; Kidd, G.J.; Chang, A.; Rudick, R.; Fisher, E.; Trapp, B.D. Imaging co-relates of decreased axonal Na+/K+ ATPase in chronic multiple sclerosis lesions. Ann. Neurol. 2008, 63, 428–435. [Google Scholar] [CrossRef] [PubMed]
  144. Stys, P.K.; Waxman, S.G.; Ransom, B.R. Ionic mechanisms of anoxic injury in mammalian CNS white matter: Role of Na+ channels and Na (+)-Ca2+ exchanger. J. Neurosci. 1992, 12, 430–439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Mathur, D.; Riffo-Campos, A.L.; Castillo, J.; Haines, J.D.; Vidaurre, O.G.; Zhang, F.; Coret-Ferrer, F.; Casaccia, P.; Casanova, B.; Lopez-Rodas, G. Bioenergetic failure in rat oligodendrocyte progenitor cells treated with cerebrospinal fluid derived from multiple sclerosis patients. Front. Cell. Neurosci. 2017, 11, 209. [Google Scholar] [CrossRef]
  146. Mathur, D.; María-Lafuente, E.; Ureña-Peralta, J.R.; Sorribes, L.; Hernández, A.; Casanova, B.; López-Rodas, G.; Coret-Ferrer, F.; Burgal-Marti, M. Disturbed glucose metabolism in rat neurons exposed to cerebrospinal fluid obtained from multiple sclerosis subjects. Brain Sci. 2017, 8, 1. [Google Scholar] [CrossRef] [Green Version]
  147. Waxman, S.G. Axonal dysfunction in chronic multiple sclerosis: Meltdown in the membrane. Ann. Neurol. 2008, 63, 411–413. [Google Scholar] [CrossRef] [PubMed]
  148. Zhu, X.H.; Qiao, H.; Du, F.; Xiong, Q.; Liu, X.; Zhang, X.; Ugurbil, K.; Chen, W. Quantitative imaging of energy expenditure in human brain. Neuroimage 2012, 60, 2107–2117. [Google Scholar] [CrossRef] [Green Version]
  149. Zaaraoui, W.; Konstandin, S.; Audoin, B.; Nagel, A.M.; Rico, A.; Malikova, I.; Soulier, E.; Viout, P.; Confort-Gouny, S.; Cozzone, P.J.; et al. Distribution of brain sodium accumulation correlates with disability in multiple sclerosis: A cross-sectional 23Na MR imaging study. Radiology 2012, 264, 859–867. [Google Scholar] [CrossRef]
  150. Paling, D.; Solanky, B.S.; Riemer, F.; Tozer, D.J.; Wheeler-Kingshott, C.A.; Kapoor, R.; Golay, X.; Miller, D.H. Sodium accumulation is associated with disability and a progressive course in multiple sclerosis. Brain 2013, 136, 2305–2317. [Google Scholar] [CrossRef]
  151. Petracca, M.; Fleysher, L.; Oesingmann, N.; Inglese, M. Sodium MRI of multiple sclerosis. NMR Biomed. 2016, 29, 153–161. [Google Scholar] [CrossRef] [Green Version]
  152. Eisele, P.; Konstandin, S.; Szabo, K.; Ebert, A.; Roßmanith, C.; Paschke, N.; Kerschensteiner, M.; Platten, M.; Schoenberg, S.O.; Schad, L.R.; et al. Temporal evolution of acute multiple sclerosis lesions on serial sodium (23Na) MRI. Mult. Scler. Relat. Disord. 2019, 29, 48–54. [Google Scholar] [CrossRef]
  153. Huhn, K.; Engelhorn, T.; Linker, R.A.; Nagel, A.M. Potential of sodium MRI as a biomarker for neurodegeneration and neuroinflammation in multiple sclerosis. Front. Neurol. 2019, 10, 84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Brebner, J.A.; Stockley, R.A. Polyclonal free light chains: A biomarker of inflammatory disease or treatment target? F1000 Med. Rep. 2013, 5, 4. [Google Scholar] [CrossRef] [PubMed]
  155. Presslauer, S.; Milosavljevic, D.; Brücke, T.; Bayer, P.; Hübl, W. Elevated levels of kappa free light chains in CSF support the diagnosis of multiple sclerosis. J. Neurol. 2008, 255, 1508–1514. [Google Scholar] [CrossRef] [PubMed]
  156. Rinker, J.R., 2nd; Trinkaus, K.; Cross, A.H. Elevated CSF free kappa light chains correlate with disability prognosis in multiple sclerosis. Neurology 2006, 67, 1288–1290. [Google Scholar] [CrossRef]
  157. Villar, L.M.; Espino, M.; Costa-Frossard, L.; Muriel, A.; Jimenez, J.; Alvarez-Cermeño, J.C. High levels of cerebrospinal fluid free kappa chains predict conversion to multiple sclerosis. Clin. Chim. Acta 2012, 413, 1813–1816. [Google Scholar] [CrossRef]
  158. Arneth, B.; Birklein, F. High sensitivity of free lambda and free kappa light chains for detection of intrathecal immunoglobulin synthesis in cerebrospinal fluid. Acta Neurol. Scand. 2009, 119, 39–44. [Google Scholar] [CrossRef]
  159. Brundin, L.; Morcos, E.; Olsson, T.; Wiklund, N.P.; Andersson, M. Increased intrathecal nitric oxide formation in multiple sclerosis, cerebrospinal fluid nitrite as activity marker. Eur. J. Neurol. 1999, 6, 585–590. [Google Scholar] [CrossRef]
  160. Danilov, A.I.; Andersson, M.; Bavand, N.; Wiklund, N.P.; Olsson, T.; Brundin, L. Nitric oxide metabolite determinations reveal continuous inflammation in multiple sclerosis. J. Neuroinmunol. 2003, 136, 112–118. [Google Scholar] [CrossRef]
  161. Brown, G.C.; Bal-Price, A. Inflammatory neurodegeneration mediated by nitric oxide, glutamate, and mitochondria. Mol. Neurobiol. 2003, 27, 325–355. [Google Scholar] [CrossRef]
  162. Sellebjerg, F.; Giovannoni, G.; Hand, A.; Madsen, H.O.; Jensen, C.V.; Garred, P. Cerebrospinal fluid levels of nitric oxide metabolites predict response to methylprednisolone treatment in multiple sclerosis and optic neuritis. J. Neuroinmunol. 2002, 125, 198–203. [Google Scholar] [CrossRef]
  163. Vladimirova, O.; Lu, F.M.; Shawver, L.; Kalman, B. The activation of protein kinase C induces higher production of reactive oxygen species by mononuclear cells in patients with multiple sclerosis than in controls. Inflamm. Res. 1999, 48, 412–416. [Google Scholar] [CrossRef]
  164. Diestel, A.; Akta, S.O.; Hackel, D.; Hake, I.; Meier, S.; Raine, C.S.; Nitsch, R.; Zipp, F.; Ullrich, O. Activation of microglial poly (ADP-ribose)-polymerase-1 by cholesterol breakdown products during neuroinflammation: A link between demyelination and neuronal damage. J. Exp. Med. 2003, 198, 1729–1740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Lucchinetti, C.F.; Bruck, W.; Rodriguez, M.; Lassmann, H. Distinct patterns of multiple sclerosis pathology indicate heterogeneity on pathogenesis. Brain Pathol. 1996, 6, 259–274. [Google Scholar] [CrossRef] [PubMed]
  166. Ozawa, K.; Suchanek, G. Patterns of oligodendroglia pathology in multiple sclerosis. Brain 1994, 117, 1311–1322. [Google Scholar] [CrossRef]
  167. Sun, M.; Liu, N.; Xie, Q.; Li, X.; Sun, J.; Wang, H.; Wang, M.A. candidate biomarker of glial fibrillary acidic protein in CSF and blood in differentiating multiple sclerosis and its subtypes: A systematic review and meta-analysis. Mult. Scler. Relat. Disord. 2021, 51, 102870. [Google Scholar] [CrossRef] [PubMed]
  168. Petzold, A.; Eikelenboom, M.J.; Gveric, D.; Keir, G.; Chapman, M.; Lazeron, R.H.; Cuzner, M.L.; Polman, C.H.; Uitdehaag, B.M.; Thompson, E.J. Markers for different glial cell responses in multiple sclerosis: Clinical and pathological correlations. Brain 2002, 125, 1462–1473. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Misu, T.; Takahashi, T.; Nakashima, I.; Fujihara, K. Biomarkers in Neuromyelitis Optica. Brain Nerve 2012, 64, 525–535. [Google Scholar]
  170. Takano, R.; Misu, T.; Takahashi, T.; Sato, S.; Fujihara, K.; Itoyama, Y. Astrocytic damage is far more severe than demyelination in NMO: A clinical CSF biomarker study. Neurology 2010, 75, 208–216. [Google Scholar] [CrossRef]
  171. Flanagan, E.P.; Cabre, P.; Weinshenker, B.G.; Sauver, J.S.; Jacobson, D.J.; Majed, M.; Lennon, V.A.; Lucchinetti, C.F.; McKeon, A.; Matiello, M.; et al. Epidemiology of aquaporin-4 autoimmunity and neuromyelitis optica spectrum. Ann. Neurol. 2016, 79, 775–783. [Google Scholar] [CrossRef]
  172. McCreary, M.; Mealy, M.A.; Wingerchuk, D.M.; Levy, M.; DeSena, A.; Greenberg, B.M. Updated diagnostic criteria for neuromyelitis optica spectrum disorder: Similar outcomes of previously separate cohorts. Mult. Scler. J. Exp. Transl. Clin. 2018, 4, 2055217318815925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Paul, F.; Jarius, S.; Aktas, O.; Bluthner, M.; Bauer, O.; Appelhans, H.; Franciotta, D.; Bergamaschi, R.; Littleton, E.; Palace, J.; et al. Antibody to aquaporin 4 in the diagnosis of neuromyelitis optica. PLoS Med. 2007, 4, e133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Schwenkenbecher, P.; Wurster, U.; Konen, F.F.; Gingele, S.; Sühs, K.W.; Wattjes, M.P.; Stangel, M.; Skripuletz, T. Impact of the McDonald Criteria 2017 on Early Diagnosis of Relapsing-Remitting Multiple Sclerosis. Front. Neurol. 2019, 10, 188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Representative images of MS lesions in the CNS obtained by the magnetic resonance techniques (MRI) most used for diagnosis in daily clinical practice. (A) Brain MRI T2-weighted. (B) Brain MRI T1-weighted. (C) Brain MRI T1-weighted after gadolinium administration. (D) Brain MRI FLAIR-weighted. (E) Spinal cord MRI STIR sequence.
Figure 1. Representative images of MS lesions in the CNS obtained by the magnetic resonance techniques (MRI) most used for diagnosis in daily clinical practice. (A) Brain MRI T2-weighted. (B) Brain MRI T1-weighted. (C) Brain MRI T1-weighted after gadolinium administration. (D) Brain MRI FLAIR-weighted. (E) Spinal cord MRI STIR sequence.
Ijms 22 10323 g001
Table 1. Classification and clinical uses of biomarkers.
Table 1. Classification and clinical uses of biomarkers.
BiomarkersDescription
PredictiveRisk to Develop MS
DiagnosticFast Interpretation of Pathological State of MS
PrognosisOutcome or Course of MS
Disease-Associated ActivityDemonstration of Current MS Condition as Inflammation, Demyelination, Cognitive Dysfunction, etc.
Response to TreatmentPredict Response to Therapy in MS Patients
Table 2. Radiological biomarkers and biomarkers identified in CSF and serum samples and their standard clinical use in MS.
Table 2. Radiological biomarkers and biomarkers identified in CSF and serum samples and their standard clinical use in MS.
Predictive BiomarkersDiagnostic
Biomarkers
Prognostic
Biomarker
Biomarkers of Dysfunction and Pathology
Epstein–Barr Virus (EBV) [26,27,28]Radiological
Biomarkers [29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54]
Chitinase-3-Like-1 (CHI3L1)
[55,56,57]
Immunological Dysfunction
Cytokines [58,59,60,61,62,63,64]
Human Herpesvirus Type-6 (HHV-6) [65,66,67,68,69,70]Oligoclonal Bands (IgG) and (IgM)
[71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94]
Neurofilaments (NF) [95,96,97,98,99,100,101,102,103,104,105]Demyelination
Myelin Basic Protein (MBP) [106,107,108,109,110,111]
Anti-MOG and
Anti-MBP Antibodies
[112,113,114,115]
Immunoglobulin G
Index (IgG Index) [116,117]
miRNAs [118,119,120,121]Axonal Damage
Neurofilaments (NF) [122,123,124,125,126,127]
Microtubule-Associated Protein Tau [128,129,130,131,132,133]
N-acetylaspartate (NAA) [134,135,136,137]
Amyloid-precursor protein (APP) [138,139,140,141,142]
Ionic Imbalance [143,144,145,146,147,148,149,150,151,152,153]
Kappa Free (KFLC) and Lambda Free Light Chains (LFLC) [154,155,156,157,158] Glial activation/dysfunction
Nitric Oxide (NO) [159,160,161,162]
Reactive Oxygen Species (ROS) [163,164]
Glial Fibrillary Acidic Protein (GFAP) [165,166,167,168,169,170]
S100 Calcium-Binding Protein B (S-100B) [168]
Anti-Aquaporin-4 (AQP4) Antibodies
[171,172,173]
Table 3. Currently imaging biomarkers used in MS, lesions that are mainly detected, and utility and limitations of each technique.
Table 3. Currently imaging biomarkers used in MS, lesions that are mainly detected, and utility and limitations of each technique.
Imaging
Biomarker
Lesion
Category
ReferencesUtility in MSLimitations
T2-WeightedPeriventricular Juxtacortical[29]Indicate Demyelination and
Inflammation
Variations in the MR Images which is Due to Variable
Radiofrequency Response in the Brain. Background Noise Makes Difficult to Separate Lesions from White Matter
T1-WeightedGd-Enhancing Lesions and Spinal Cord Lesions[31,32]Shows About Axonal Loss and BBB
Disruption
Need of Reproducible and Reliable Method to Quantify Brain Atrophy
MTRGd-Enhancing Demyelinated
Remyelinated and Necrotic Lesions
[38,39]Indicate Changes in Myelin Composition and Axonal LossMeasurements are Not Absolute and Results Vary
Repeatedly as a Function of the Shape, Bandwidth and Frequency of MS Pulse so More Errors Occur
DWI and DTIDemyelinating and Periventricular Lesions[41,42,43]Demonstrates about Demyelination and Axonal LossLack of Pathology Specificity. Required Variable Samples According to Different Type of Lesions and Different
Locations of the Brain
MRSPeriventricular and Cortical
Lesions
[49,50]Assess Measure Biochemicals as NAA, GABA, etc. which Reflect Axonal Damage and Shows Abnormalities in Pathology of MS
OCTOptic Nerve Lesions[51,52]Used to Measure RNFL Thickness and Macular Volume also Gives Data about
Axonal Loss, Neurodegeneration
Movement of Patient Can Diminish the Quality of Image
PETPeriventricular Necrotic
Juxtacortical and
Gd-enhancing Lesions
[53,54]Assess Inflammation in Cortex, NAWM and Gives Information about
Clinical Severity
Invasive and Expensive Tool Not Available all the Time. During Acquisition, if Patient Moves the Activity Will Blur Over Brain Structures. Resolution Will be Degraded and Interpretation of Results Will be Impossible
MTR: Magnetization Transfer Ratio, DWI: Diffusion-Weighted Imaging, DTI: Diffusion Tensor Imaging, PET: Positron Emission Tomography, OCT: Optical Coherence Tomography, MRS: Magnetic Resonance Spectroscopy; RNFL: retinal nerve fiber layer.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mathur, D.; Mishra, B.K.; Rout, S.; Lopez-Iranzo, F.J.; Lopez-Rodas, G.; Vallamkondu, J.; Kandimalla, R.; Casanova, B. Potential Biomarkers Associated with Multiple Sclerosis Pathology. Int. J. Mol. Sci. 2021, 22, 10323. https://doi.org/10.3390/ijms221910323

AMA Style

Mathur D, Mishra BK, Rout S, Lopez-Iranzo FJ, Lopez-Rodas G, Vallamkondu J, Kandimalla R, Casanova B. Potential Biomarkers Associated with Multiple Sclerosis Pathology. International Journal of Molecular Sciences. 2021; 22(19):10323. https://doi.org/10.3390/ijms221910323

Chicago/Turabian Style

Mathur, Deepali, Bikash Kumar Mishra, Soumyashree Rout, Francisco Jose Lopez-Iranzo, Gerardo Lopez-Rodas, Jayalakshmi Vallamkondu, Ramesh Kandimalla, and Bonaventura Casanova. 2021. "Potential Biomarkers Associated with Multiple Sclerosis Pathology" International Journal of Molecular Sciences 22, no. 19: 10323. https://doi.org/10.3390/ijms221910323

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop