The Molecular and Cellular Basis for Multiple Sclerosis 2020

A special issue of Cells (ISSN 2073-4409). This special issue belongs to the section "Cellular Pathology".

Deadline for manuscript submissions: closed (30 June 2020) | Viewed by 44948

Special Issue Editors


E-Mail Website
Guest Editor
Neurology Department, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Universitätsklinikum Essen, Hufelandstr. 55, 45147 Essen, Germany
Interests: neuroimmunology; multiple sclerosis (clinical/experimental); stroke (clinical/experimental); thromboinflammation; neuroprotection; neuroimaging
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Neurology Department, Center for Translational Neuro- and Behavioral Sciences (C-TNBS), Universitätsklinikum Essen, Hufelandstr. 55, 45147 Essen, Germany
Interests: neuroimmunology; multiple sclerosis (clinical/experimental); animal models; glial cells; biomarkers in MS; neuroprotection; neuroimaging
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

Multiple sclerosis (MS) is a chronic inflammatory and demyelinating disease. Despite extensive research, the question of whether it is triggered by an initial event outside the CNS (outside–in hypothesis) or whether it is a CNS-intrinsic event (inside–out hypothesis) is still not resolved. The discovery of several molecular and cellular mechanisms has provided valuable insight into the disease mechanism of multiple sclerosis (MS). The former concept that inflammation is only confined to white matter lesions and that autoreactive CD4+ T cells are the exclusive disease contributors no longer holds true. MS is a systemic disease that affects both the grey and white matter of the central nervous system (CNS). Inflammatory and degenerative mechanisms take place at the same time, and their balance probably determines a relapsing–remitting or a progressive disease course. It remains elusive whether primary progressive MS is a disease entity of its own or merely a disease phenotype that lacks superimposed high-grade inflammatory mechanisms. The same question is valid for highly active MS with tumefactive lesions, and the question arises whether there is a molecular key that determines an excessive or a more subtle immune response. Recent research reveals that it does not seem to be a single mechanism, and that there is a need to discover and classify inflammatory molecular and cellular patterns. Understanding these patterns and their dynamics in association with clinical and imaging data will be of utmost importance in deciphering disease mechanisms. In contrast to inflammation, our knowledge about axonal and neural degeneration is quite more restricted. It has been shown that mechanisms underlying neurodegeneration, at least in the long term, proceed autonomously and may be the central pathological feature. The elucidation of emerging cellular and molecular pathways and their relationship to demyelination and inflammation will herald a breakthrough in our understanding of the disease and in developing targeted therapies for neurodegeneration. Moreover, the significant progress made in analyzing genetic risk factors by using genome-wide association studies may not only help to shed light on individual susceptibility and environmental risk factors to develop the disease but also to unravel supervisory roles at the genetic and epigenetic level. More than 150 single nucleotide polymorphisms (SNP) outside the human leucocyte antigen (HLA) system have been discovered and mostly implicate immune genes. For most of the genes located at those SNPs, it must first be verified whether they are functionally relevant at all.

The inclusion of the oligoclonal bands (OCBs) in the new 2017 revised McDonald criteria supports the concept of early diagnosis and treatment but also underlines the need of further molecular and cellular predictors in MS. The current spotlight is on the soluble neurofilament light chain as a disease marker with the capability to follow the footsteps of OCBs. Indeed, there is an urgent need for molecular and cellular biomarkers not only to evaluate MS therapies but also to fill the gap between relapses and MRI lesions. A biomarker that exactly displays the inflammatory and degenerative components of the disease will enable an exactly adapted therapy without losing time in treatment optimization and without hesitation in the treatment of special conditions like radiologically isolated syndrome.

It is fascinating to see how many new drugs have been approved for MS in the last years. Changing the focus to B cells or the idea to “grow a healthier” or more anti-inflammatory immune system after cell depletion are new treatment approaches, but molecular and cellular mechanisms that may explain their effects are lacking. Research focused on the mode of action of MS drugs is of great interest, since it is inevitably associated with the unveiling of disease mechanisms.

This Special Issue is dedicated to the description of pathological cellular and molecular mechanisms, the identification of the fundamental basis of the disease, and the development of molecular interventions to prevent or treat them towards a molecular medicinal perspective of MS.

Prof. Dr. Christoph Kleinschnitz
Prof. Dr. Sven G. Meuth
Dr. Refik Pul
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Cells is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • multiple sclerosis
  • demyelination
  • remyelination
  • inflammation
  • neurodegeneration
  • biomarker
  • disease mechanism
  • mode of action of MS drugs

Published Papers (9 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

15 pages, 554 KiB  
Article
HLA Class II Genotype Does Not Affect the Myelin Responsiveness of Multiple Sclerosis Patients
by Judith Derdelinckx, Irene Nkansah, Naomi Ooms, Laura Van Bruggen, Marie-Paule Emonds, Liesbeth Daniëls, Tatjana Reynders, Barbara Willekens, Patrick Cras, Zwi N. Berneman and Nathalie Cools
Cells 2020, 9(12), 2703; https://doi.org/10.3390/cells9122703 - 17 Dec 2020
Viewed by 2556
Abstract
Background: When aiming to restore myelin tolerance using antigen-specific treatment approaches in MS, the wide variety of myelin-derived antigens towards which immune responses are targeted in multiple sclerosis (MS) patients needs to be taken into account. Uncertainty remains as to whether the myelin [...] Read more.
Background: When aiming to restore myelin tolerance using antigen-specific treatment approaches in MS, the wide variety of myelin-derived antigens towards which immune responses are targeted in multiple sclerosis (MS) patients needs to be taken into account. Uncertainty remains as to whether the myelin reactivity pattern of a specific MS patient can be predicted based upon the human leukocyte antigen (HLA) class II haplotype of the patient. Methods: In this study, we analyzed the reactivity towards myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP) and proteolipid protein (PLP) peptides using direct interferon (IFN)-γ enzyme-linked immune absorbent spot (ELISPOT). Next, the HLA class II haplotype profile was determined by next-generation sequencing. In doing so, we aimed to evaluate the possible association between the precursor frequency of myelin-reactive T cells and the HLA haplotype. Results: Reactivity towards any of the analyzed peptides could be demonstrated in 65.0% (13/20) of MS patients and in 60.0% (6/10) of healthy controls. At least one of the MS risk alleles HLA-DRB1*15:01, HLA-DQA1*01:02 and HLA-DQB1*06:02 was found in 70.0% (14/20) of patients and in 20.0% (2/10) of healthy controls. No difference in the presence of a myelin-specific response, nor in the frequency of myelin peptide-reactive precursor cells could be detected among carriers and non-carriers of these risk alleles. Conclusion: No association between HLA haplotype and myelin reactivity profile was present in our study population. This complicates the development of antigen-specific treatment approaches and implies the need for multi-epitope targeting in an HLA-unrestricted manner to fully address the wide variation in myelin responses and HLA profiles in a heterogeneous group of MS patients. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

13 pages, 2104 KiB  
Article
Gas6 Induces Myelination through Anti-Inflammatory IL-10 and TGF-β Upregulation in White Matter and Glia
by Salman Goudarzi, Shannon E. Gilchrist and Sassan Hafizi
Cells 2020, 9(8), 1779; https://doi.org/10.3390/cells9081779 - 26 Jul 2020
Cited by 21 | Viewed by 3431
Abstract
The Gas6–TAM (Tyro3, Axl, Mer) ligand–receptor system is believed to promote central nervous system (CNS) (re)myelination and glial cell development. An additional important function of Gas6–TAM signalling appears to be the regulation of immunity and inflammation, which remains to be fully elucidated in [...] Read more.
The Gas6–TAM (Tyro3, Axl, Mer) ligand–receptor system is believed to promote central nervous system (CNS) (re)myelination and glial cell development. An additional important function of Gas6–TAM signalling appears to be the regulation of immunity and inflammation, which remains to be fully elucidated in the CNS. Here, we characterised the expression of TAM receptors and ligands in individual CNS glial cell types, observing high expression of Gas6 and the TAM receptors, Mer and Axl, in microglia, and high expression of Tyro3 in astrocytes. We also investigated the effect of Gas6 on the inflammatory cytokine response in the optic nerve and in mixed glial cell cultures from wildtype and single TAM receptor knockout mice. In wildtype and Mer-deficient cultures, Gas6 significantly stimulated the expression of the anti-inflammatory/pro-repair cytokines interleukin 10 (IL-10) and transforming growth factor β (TGF-β), whereas this effect was absent in either Tyro3 or Axl knockout cultures. Furthermore, Gas6 caused upregulation of myelin basic protein (MBP) expression in optic nerves, which was blocked by a neutralising antibody against IL-10. In conclusion, our data show that microglia are both a major source of Gas6 as well as an effector of Gas6 action in the CNS through the upregulation of anti-inflammatory and pro-repair mediators. Furthermore, the presence of both Axl and Tyro3 receptors appears to be necessary for these effects of Gas6. In addition, IL-10, alongside suppressing inflammation and immunity, mediates the pro-myelinating mechanism of Gas6 action in the optic nerve. Therefore, Gas6 may present an attractive target for novel therapeutic interventions for demyelinating as well as neuroinflammatory disorders of the CNS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

16 pages, 2214 KiB  
Article
The Impact of Immunomodulatory Treatment on Kappa Free Light Chains as Biomarker in Neuroinflammation
by Franz Felix Konen, Ulrich Wurster, Torsten Witte, Konstantin Fritz Jendretzky, Stefan Gingele, Hayrettin Tumani, Kurt-Wolfram Sühs, Martin Stangel, Philipp Schwenkenbecher and Thomas Skripuletz
Cells 2020, 9(4), 842; https://doi.org/10.3390/cells9040842 - 31 Mar 2020
Cited by 23 | Viewed by 2910
Abstract
Background: Kappa free light chains (KFLC) are a promising new biomarker to detect neuroinflammation. Still, the impact of pre-analytical effects on KFLC concentrations was not investigated. Methods: KFLC concentrations were measured in serum and cerebrospinal fluid (CSF) of patients with a newly diagnosed [...] Read more.
Background: Kappa free light chains (KFLC) are a promising new biomarker to detect neuroinflammation. Still, the impact of pre-analytical effects on KFLC concentrations was not investigated. Methods: KFLC concentrations were measured in serum and cerebrospinal fluid (CSF) of patients with a newly diagnosed multiple sclerosis (MS) or clinically isolated syndrome (CIS) before (n = 42) or after therapy with high-dose methylprednisolone (n = 65). In prospective experiments, KFLC concentrations were analyzed in the same patients in serum before and after treatment with high-dose methylprednisolone (n = 16), plasma exchange (n = 12), immunoadsorption (n = 10), or intravenous immunoglobulins (n = 10). In addition, the influence of storage time, sample method, and contamination of CSF with blood were investigated. Results: Patients diagnosed with MS/CIS and treated with methylprednisolone showed significantly lower KFLC concentrations in serum as untreated patients. Repeated longitudinal investigations revealed that serum KFLC concentrations continuously decreased after each application of methylprednisolone. In contrast, other immune therapies and further pre-analytical conditions did not influence KFLC concentrations. Conclusion: Our results show prominent effects of steroids on KFLC concentrations. In contrast, various other pre-analytical conditions did not influence KFLC concentrations, indicating the stability of this biomarker. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

23 pages, 7721 KiB  
Article
The Rare IL22RA2 Signal Peptide Coding Variant rs28385692 Decreases Secretion of IL-22BP Isoform-1, -2 and -3 and Is Associated with Risk for Multiple Sclerosis
by Paloma Gómez-Fernández, Aitzkoa Lopez de Lapuente Portilla, Ianire Astobiza, Jorge Mena, Andoni Urtasun, Vivian Altmann, Fuencisla Matesanz, David Otaegui, Elena Urcelay, Alfredo Antigüedad, Sunny Malhotra, Xavier Montalban, Tamara Castillo-Triviño, Laura Espino-Paisán, Orhan Aktas, Mathias Buttmann, Andrew Chan, Bertrand Fontaine, Pierre-Antoine Gourraud, Michael Hecker, Sabine Hoffjan, Christian Kubisch, Tania Kümpfel, Felix Luessi, Uwe K. Zettl, Frauke Zipp, Iraide Alloza, Manuel Comabella, Christina M. Lill and Koen Vandenbroeckadd Show full author list remove Hide full author list
Cells 2020, 9(1), 175; https://doi.org/10.3390/cells9010175 - 10 Jan 2020
Cited by 4 | Viewed by 4638
Abstract
The IL22RA2 locus is associated with risk for multiple sclerosis (MS) but causative variants are yet to be determined. In a single nucleotide polymorphism (SNP) screen of this locus in a Basque population, rs28385692, a rare coding variant substituting Leu for Pro at [...] Read more.
The IL22RA2 locus is associated with risk for multiple sclerosis (MS) but causative variants are yet to be determined. In a single nucleotide polymorphism (SNP) screen of this locus in a Basque population, rs28385692, a rare coding variant substituting Leu for Pro at position 16 emerged significantly (p = 0.02). This variant is located in the signal peptide (SP) shared by the three secreted protein isoforms produced by IL22RA2 (IL-22 binding protein-1(IL-22BPi1), IL-22BPi2 and IL-22BPi3). Genotyping was extended to a Europe-wide case-control dataset and yielded high significance in the full dataset (p = 3.17 × 10−4). Importantly, logistic regression analyses conditioning on the main known MS-associated SNP at this locus, rs17066096, revealed that this association was independent from the primary association signal in the full case-control dataset. In silico analysis predicted both disruption of the alpha helix of the H-region of the SP and decreased hydrophobicity of this region, ultimately affecting the SP cleavage site. We tested the effect of the p.Leu16Pro variant on the secretion of IL-22BPi1, IL-22BPi2 and IL-22BPi3 and observed that the Pro16 risk allele significantly lowers secretion levels of each of the isoforms to around 50%–60% in comparison to the Leu16 reference allele. Thus, our study suggests that genetically coded decreased levels of IL-22BP isoforms are associated with augmented risk for MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

18 pages, 3629 KiB  
Article
Multiple Sclerosis CD49d+CD154+ As Myelin-Specific Lymphocytes Induced During Remyelination
by Paweł Piatek, Magdalena Namiecinska, Małgorzata Domowicz, Marek Wieczorek, Sylwia Michlewska, Mariola Matysiak, Natalia Lewkowicz, Maciej Tarkowski and Przemysław Lewkowicz
Cells 2020, 9(1), 15; https://doi.org/10.3390/cells9010015 - 19 Dec 2019
Cited by 8 | Viewed by 4026
Abstract
Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS) mediated by autoreactive lymphocytes. The role of autoreactive lymphocytes in the CNS demyelination is well described, whereas very little is known about their role in remyelination during MS remission. [...] Read more.
Multiple sclerosis (MS) is a demyelinating autoimmune disease of the central nervous system (CNS) mediated by autoreactive lymphocytes. The role of autoreactive lymphocytes in the CNS demyelination is well described, whereas very little is known about their role in remyelination during MS remission. In this study, we identified a new subpopulation of myelin-specific CD49d+CD154+ lymphocytes presented in the peripheral blood of MS patients during remission, that proliferated in vitro in response to myelin peptides. These lymphocytes possessed the unique ability to migrate towards maturing oligodendrocyte precursor cells (OPCs) and synthetize proinflammatory chemokines/cytokines. The co-culture of maturing OPCs with myelin-specific CD49d+CD154+ lymphocytes was characterized by the increase in proinflammatory chemokine/cytokine secretion that was not only a result of their cumulative effect of what OPCs and CD49d+CD154+ lymphocytes produced alone. Moreover, maturing OPCs exposed to exogenous myelin peptides managed to induce CD40-CD154-dependent CD49d+CD154+ lymphocyte proliferation. We confirmed, in vivo, the presence of CD49d+CD154+ cells close to maturating OPCs and remyelinating plaque during disease remission in the MS mouse model (C57Bl/6 mice immunized with MOG35-55) by immunohistochemistry. Three weeks after an acute phase of experimental autoimmune encephalomyelitis, CD49d+/CD154+ cells were found to be co-localized with O4+ cells (oligodendrocyte progenitors) in the areas of remyelination identified by myelin basic protein (MBP) labelling. These data suggested that myelin-specific CD49d+CD154+ lymphocytes present in the brain can interfere with remyelination mediated by oligodendrocytes probably as a result of establishing proinflammatory environment. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

22 pages, 3942 KiB  
Article
MS CD49d+CD154+ Lymphocytes Reprogram Oligodendrocytes into Immune Reactive Cells Affecting CNS Regeneration
by Paweł Piatek, Magdalena Namiecinska, Małgorzata Domowicz, Patrycja Przygodzka, Marek Wieczorek, Sylwia Michlewska, Natalia Lewkowicz, Maciej Tarkowski and Przemysław Lewkowicz
Cells 2019, 8(12), 1508; https://doi.org/10.3390/cells8121508 - 25 Nov 2019
Cited by 7 | Viewed by 3752
Abstract
The critical aspect in multiple sclerosis (MS) progression involves insufficient regeneration of CNS resulting from deficient myelin synthesis by newly generated oligodendrocytes (OLs). Although many studies have focused on the role of autoreactive lymphocytes in the inflammatory-induced axonal loss, the problem of insufficient [...] Read more.
The critical aspect in multiple sclerosis (MS) progression involves insufficient regeneration of CNS resulting from deficient myelin synthesis by newly generated oligodendrocytes (OLs). Although many studies have focused on the role of autoreactive lymphocytes in the inflammatory-induced axonal loss, the problem of insufficient remyelination and disease progression is still unsolved. To determine the effect of myelin-specific lymphocytes on OL function in MS patients and in a mouse model of MS, we cultured myelin induced MS CD49d+CD154+ circulating lymphocytes as well as Experimental Autoimmune Encephalomyelitis (EAE) mouse brain-derived T and memory B cells with maturing oligodendrocyte precursor cells (OPCs). We found that myelin-specific CD49d+CD154+ lymphocytes affected OPC maturation toward formation of immune reactive OLs. Newly generated OLs were characterized by imbalanced myelin basic protein (MBP) and proteolipid protein (PLP) production as well as proinflammatory chemokine/cytokine synthesis. The analysis of cellular pathways responsible for OL reprogramming revealed that CD49d+CD154+ lymphocytes affected miRNA synthesis by dysregulation of polymerase II activity. miR-665 and ELL3 turned out to be the main targets of MS myelin-specific lymphocytes. Neutralization of high intracellular miR-665 concentration restored miRNA and MBP/PLP synthesis. Together, these data point to new targets for therapeutic intervention promoting CNS remyelination. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Graphical abstract

Review

Jump to: Research

21 pages, 1089 KiB  
Review
The Role of MicroRNAs in Repair Processes in Multiple Sclerosis
by Conor P. Duffy and Claire E. McCoy
Cells 2020, 9(7), 1711; https://doi.org/10.3390/cells9071711 - 16 Jul 2020
Cited by 25 | Viewed by 5562
Abstract
Multiple sclerosis (MS) is an autoimmune disorder characterised by demyelination of central nervous system neurons with subsequent damage, cell death and disability. While mechanisms exist in the CNS to repair this damage, they are disrupted in MS and currently there are no treatments [...] Read more.
Multiple sclerosis (MS) is an autoimmune disorder characterised by demyelination of central nervous system neurons with subsequent damage, cell death and disability. While mechanisms exist in the CNS to repair this damage, they are disrupted in MS and currently there are no treatments to address this deficit. In recent years, increasing attention has been paid to the influence of the small, non-coding RNA molecules, microRNAs (miRNAs), in autoimmune disorders, including MS. In this review, we examine the role of miRNAs in remyelination in the different cell types that contribute to MS. We focus on key miRNAs that have a central role in mediating the repair process, along with several more that play either secondary or inhibitory roles in one or more aspects. Finally, we consider the current state of miRNAs as therapeutic targets in MS, acknowledging current challenges and potential strategies to overcome them in developing effective novel therapeutics to enhance repair mechanisms in MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

17 pages, 977 KiB  
Review
The Meaning of Immune Reconstitution after Alemtuzumab Therapy in Multiple Sclerosis
by Simona Rolla, Alessandro Maglione, Stefania Federica De Mercanti and Marinella Clerico
Cells 2020, 9(6), 1396; https://doi.org/10.3390/cells9061396 - 03 Jun 2020
Cited by 16 | Viewed by 4710
Abstract
Alemtuzumab is a monoclonal antibody that binds to CD52, a protein present on the surface of mature lymphocytes, but not on the stem cells from which these lymphocytes are derived. It is currently used as an immune reconstitution therapy in patients with relapsing–remitting [...] Read more.
Alemtuzumab is a monoclonal antibody that binds to CD52, a protein present on the surface of mature lymphocytes, but not on the stem cells from which these lymphocytes are derived. It is currently used as an immune reconstitution therapy in patients with relapsing–remitting multiple sclerosis. Alemtuzumab treatment is an intermittent infusion that induces long-term remission of Multiple Sclerosis also in the treatment-free period. After the robust T and B cell depletion induced by alemtuzumab, the immune system undergoes radical changes during its reconstitution. In this review, we will discuss the current knowledge on the reconstitution of the lymphocyte repertoire after alemtuzumab treatment and how it could affect the development of side effects, which led to its temporary suspension by the European Medical Agency. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

20 pages, 1057 KiB  
Review
T Helper Cells: The Modulators of Inflammation in Multiple Sclerosis
by Martina Kunkl, Simone Frascolla, Carola Amormino, Elisabetta Volpe and Loretta Tuosto
Cells 2020, 9(2), 482; https://doi.org/10.3390/cells9020482 - 19 Feb 2020
Cited by 121 | Viewed by 12245
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by the progressive loss of axonal myelin in several areas of the central nervous system (CNS) that is responsible for clinical symptoms such as muscle spasms, optic neuritis, and paralysis. The progress made in [...] Read more.
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by the progressive loss of axonal myelin in several areas of the central nervous system (CNS) that is responsible for clinical symptoms such as muscle spasms, optic neuritis, and paralysis. The progress made in more than one decade of research in animal models of MS for clarifying the pathophysiology of MS disease validated the concept that MS is an autoimmune inflammatory disorder caused by the recruitment in the CNS of self-reactive lymphocytes, mainly CD4+ T cells. Indeed, high levels of T helper (Th) cells and related cytokines and chemokines have been found in CNS lesions and in cerebrospinal fluid (CSF) of MS patients, thus contributing to the breakdown of the blood–brain barrier (BBB), the activation of resident astrocytes and microglia, and finally the outcome of neuroinflammation. To date, several types of Th cells have been discovered and designated according to the secreted lineage-defining cytokines. Interestingly, Th1, Th17, Th1-like Th17, Th9, and Th22 have been associated with MS. In this review, we discuss the role and interplay of different Th cell subpopulations and their lineage-defining cytokines in modulating the inflammatory responses in MS and the approved as well as the novel therapeutic approaches targeting T lymphocytes in the treatment of the disease. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis 2020)
Show Figures

Figure 1

Back to TopTop