The Molecular and Cellular Basis for Multiple Sclerosis

A special issue of Cells (ISSN 2073-4409).

Deadline for manuscript submissions: closed (15 August 2019) | Viewed by 108648

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 hold 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 determine 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 into 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 (18 papers)

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

Research

Jump to: Review, Other

21 pages, 99073 KiB  
Article
Potential of Adult Endogenous Neural Stem/Progenitor Cells in the Spinal Cord to Contribute to Remyelination in Experimental Autoimmune Encephalomyelitis
by Yuki Maeda, Nami Nakagomi, Akiko Nakano-Doi, Hiroto Ishikawa, Yoshiki Tatsumi, Yoshio Bando, Hiroo Yoshikawa, Tomohiro Matsuyama, Fumi Gomi and Takayuki Nakagomi
Cells 2019, 8(9), 1025; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8091025 - 03 Sep 2019
Cited by 23 | Viewed by 5415
Abstract
Demyelination and remyelination play pivotal roles in the pathological process of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), a well-established animal model of MS. Although increasing evidence shows that various stimuli can promote the activation/induction of endogenous neural stem/progenitor cells (NSPCs) in [...] Read more.
Demyelination and remyelination play pivotal roles in the pathological process of multiple sclerosis (MS) and experimental autoimmune encephalomyelitis (EAE), a well-established animal model of MS. Although increasing evidence shows that various stimuli can promote the activation/induction of endogenous neural stem/progenitor cells (NSPCs) in the central nervous system, the potential contributions of these cells to remyelination following inflammatory injury remain to be fully investigated. In the present study, using an adult mouse model of EAE induced by myelin oligodendrocyte glycoprotein (MOG) peptide, we investigated whether adult NSPCs in the spinal cord can lead to remyelination under inflammatory conditions. Immunohistochemistry showed that cells expressing the NSPC marker Nestin appeared after MOG peptide administration, predominantly at the sites of demyelination where abundant inflammatory cells had accumulated, whereas Nestin+ cells were rarely present in the spinal cord of PBS-treated control mice. In vitro, Nestin+ NSPCs obtained from EAE mice spinal cords could differentiate into multiple neural lineages, including neurons, astrocytes, and myelin-producing oligodendrocytes. Using the Cre–LoxP system, we established a mouse strain expressing yellow fluorescent protein (YFP) under the control of the Nestin promoter and investigated the expression patterns of YFP-expressing cells in the spinal cord after EAE induction. At the chronic phase of the disease, immunohistochemistry showed that YFP+ cells in the injured regions expressed markers for various neural lineages, including myelin-forming oligodendrocytes. These results show that adult endogenous NSPCs in the spinal cord can be subject to remyelination under inflammatory conditions, such as after EAE, suggesting that endogenous NSPCs represent a therapeutic target for MS treatment. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

17 pages, 7169 KiB  
Article
Naturally Occurring Nervonic Acid Ester Improves Myelin Synthesis by Human Oligodendrocytes
by Natalia Lewkowicz, Paweł Piątek, Magdalena Namiecińska, Małgorzata Domowicz, Radosław Bonikowski, Janusz Szemraj, Patrycja Przygodzka, Mariusz Stasiołek and Przemysław Lewkowicz
Cells 2019, 8(8), 786; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8080786 - 29 Jul 2019
Cited by 43 | Viewed by 5552
Abstract
The dysfunction of oligodendrocytes (OLs) is regarded as one of the major causes of inefficient remyelination in multiple sclerosis, resulting gradually in disease progression. Oligodendrocytes are derived from oligodendrocyte progenitor cells (OPCs), which populate the adult central nervous system, but their physiological capability [...] Read more.
The dysfunction of oligodendrocytes (OLs) is regarded as one of the major causes of inefficient remyelination in multiple sclerosis, resulting gradually in disease progression. Oligodendrocytes are derived from oligodendrocyte progenitor cells (OPCs), which populate the adult central nervous system, but their physiological capability to myelin synthesis is limited. The low intake of essential lipids for sphingomyelin synthesis in the human diet may account for increased demyelination and the reduced efficiency of the remyelination process. In our study on lipid profiling in an experimental autoimmune encephalomyelitis brain, we revealed that during acute inflammation, nervonic acid synthesis is silenced, which is the effect of shifting the lipid metabolism pathway of common substrates into proinflammatory arachidonic acid production. In the experiments on the human model of maturating oligodendrocyte precursor cells (hOPCs) in vitro, we demonstrated that fish oil mixture (FOM) affected the function of hOPCs, resulting in the improved synthesis of myelin basic protein, myelin oligodendrocyte glycoprotein, and proteolipid protein, as well as sphingomyelin. Additionally, FOM reduces proinflammatory cytokines and chemokines, and enhances fibroblast growth factor 2 (FGF2) and vascular endothelial growth factor (VEGF) synthesis by hOPCs was also demonstrated. Based on these observations, we propose that the intake of FOM rich in the nervonic acid ester may improve OL function, affecting OPC maturation and limiting inflammation. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

19 pages, 2888 KiB  
Article
Relationship between Multiple Sclerosis-Associated IL2RA Risk Allele Variants and Circulating T Cell Phenotypes in Healthy Genotype-Selected Controls
by Sophie Buhelt, Helle Bach Søndergaard, Annette Oturai, Henrik Ullum, Marina Rode von Essen and Finn Sellebjerg
Cells 2019, 8(6), 634; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8060634 - 25 Jun 2019
Cited by 15 | Viewed by 3938
Abstract
Single nucleotide polymorphisms (SNPs) in or near the IL2RA gene, that encodes the interleukin-2 (IL-2) receptor α (CD25), are associated with increased risk of immune-mediated diseases including multiple sclerosis (MS). We investigated how the MS-associated IL2RA SNPs rs2104286 and rs11256593 are associated with [...] Read more.
Single nucleotide polymorphisms (SNPs) in or near the IL2RA gene, that encodes the interleukin-2 (IL-2) receptor α (CD25), are associated with increased risk of immune-mediated diseases including multiple sclerosis (MS). We investigated how the MS-associated IL2RA SNPs rs2104286 and rs11256593 are associated with CD25 expression on T cells ex vivo by multiparameter flow cytometry in paired genotype-selected healthy controls. We observed that MS-associated IL2RA SNPs rs2104286 and rs11256593 are associated with expression of CD25 in CD4+ but not CD8+ T cells. In CD4+ T cells, carriers of the risk genotype had a reduced frequency of CD25+ TFH1 cells (p = 0.001) and an increased frequency of CD25+ recent thymic emigrant cells (p = 0.006). Furthermore, carriers of the risk genotype had a reduced surface expression of CD25 in post-thymic expanded CD4+ T cells (CD31CD45RA+), CD39+ TReg cells and in several non-follicular memory subsets. Our study found novel associations of MS-associated IL2RA SNPs on expression of CD25 in CD4+ T cell subsets. Insight into the associations of MS-associated IL2RA SNPs, as these new findings provide, offers a better understanding of CD25 variation in the immune system and can lead to new insights into how MS-associated SNPs contribute to development of MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

17 pages, 1957 KiB  
Article
CD28 Autonomous Signaling Up-Regulates C-Myc Expression and Promotes Glycolysis Enabling Inflammatory T Cell Responses in Multiple Sclerosis
by Martina Kunkl, Manolo Sambucci, Serena Ruggieri, Carola Amormino, Carla Tortorella, Claudio Gasperini, Luca Battistini and Loretta Tuosto
Cells 2019, 8(6), 575; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8060575 - 11 Jun 2019
Cited by 27 | Viewed by 4320
Abstract
The immunopathogenesis of multiple sclerosis (MS) depend on the expansion of specific inflammatory T cell subsets, which are key effectors of tissue damage and demyelination. Emerging studies evidence that a reprogramming of T cell metabolism may occur in MS, thus the identification of [...] Read more.
The immunopathogenesis of multiple sclerosis (MS) depend on the expansion of specific inflammatory T cell subsets, which are key effectors of tissue damage and demyelination. Emerging studies evidence that a reprogramming of T cell metabolism may occur in MS, thus the identification of stimulatory molecules and associated signaling pathways coordinating the metabolic processes that amplify T cell inflammation in MS is pivotal. Here, we characterized the involvement of the cluster of differentiation (CD)28 and associated signaling mediators in the modulation of the metabolic programs regulating pro-inflammatory T cell functions in relapsing-remitting MS (RRMS) patients. We show that CD28 up-regulates glycolysis independent of the T cell receptor (TCR) engagement by promoting the increase of c-myc and the glucose transporter, Glut1, in RRMS CD4+ T cells. The increase of glycolysis induced by CD28 was important for the expression of inflammatory cytokines related to T helper (Th)17 cells, as demonstrated by the strong inhibition exerted by impairing the glycolytic pathway. Finally, we identified the class 1A phosphatidylinositol 3-kinase (PI3K) as the critical signaling mediator of CD28 that regulates cell metabolism and amplify specific inflammatory T cell phenotypes in MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

12 pages, 2266 KiB  
Article
Distinct Expression of Inflammatory Features in T Helper 17 Cells from Multiple Sclerosis Patients
by Alessia Capone, Manuela Bianco, Gabriella Ruocco, Marco De Bardi, Luca Battistini, Serena Ruggieri, Claudio Gasperini, Diego Centonze, Claudio Sette and Elisabetta Volpe
Cells 2019, 8(6), 533; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8060533 - 04 Jun 2019
Cited by 14 | Viewed by 3680
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). T helper (Th) 17 lymphocytes play a role in the pathogenesis of MS. Indeed, Th17 cells are abundant in the cerebrospinal fluid and peripheral blood of MS patients and [...] Read more.
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). T helper (Th) 17 lymphocytes play a role in the pathogenesis of MS. Indeed, Th17 cells are abundant in the cerebrospinal fluid and peripheral blood of MS patients and promote pathogenesis in the mouse model of MS. To gain insight into the function of Th17 cells in MS, we tested whether Th17 cells polarized from naïve CD4 T cells of healthy donors and MS patients display different features. To this end, we analysed several parameters that typify the Th17 profile during the differentiation process of naïve CD4 T cells obtained from relapsing-remitting (RR)-MS patients (n = 31) and healthy donors (HD) (n = 28). Analysis of an array of cytokines produced by Th17 cells revealed that expression of interleukin (IL)-21, tumour necrosis factor (TNF)-β, IL-2 and IL-1R1 is significantly increased in Th17 cells derived from MS patients compared to healthy donor-derived cells. Interestingly, IL-1R1 expression is also increased in Th17 cells circulating in the blood of MS patients compared to healthy donors. Since IL-2, IL-21, TNF-β, and IL-1R1 play a crucial role in the activation of immune cells, our data indicate that high expression of these molecules in Th17 cells from MS patients could be related to their high inflammatory status. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

13 pages, 499 KiB  
Article
CD4+/CD45RO+: A Potential Biomarker of the Clinical Response to Glatiramer Acetate
by Martin Vališ, Lukáš Sobíšek, Oldřich Vyšata, Blanka Klímová, Ctirad Andrýs, Doris Vokurková, Jiří Masopust and Zbyšek Pavelek
Cells 2019, 8(5), 456; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8050456 - 15 May 2019
Viewed by 3034
Abstract
Background: Glatiramer acetate (GA) is an effective treatment for the earliest stages of multiple sclerosis (MS)—clinically isolated syndrome (CIS) or clinically definite MS (CDMS). Objective: This study aims to determine the differences in the lymphocyte population (at baseline and the course of [...] Read more.
Background: Glatiramer acetate (GA) is an effective treatment for the earliest stages of multiple sclerosis (MS)—clinically isolated syndrome (CIS) or clinically definite MS (CDMS). Objective: This study aims to determine the differences in the lymphocyte population (at baseline and the course of five years) between confirmed sustained progression (CSP) and non-CSP groups and to identify potential biomarkers among these parameters that can predict a positive response to the treatment. Methods: Twelve male and 60 female patients were included in the study. Peripheral blood samples were collected before and five years after treatment with GA. The authors compared lymphocyte parameters between the CSP and non-CSP groups by statistical analyses. Univariate and penalized logistic regression models were fitted to identify the best lymphocyte parameters at baseline and their combination for potential biomarkers. Subsequently, the ROC analysis was used to identify cut-offs for selected parameters. Results: The parameter CD4+/CD45RO+ was identified as the best single potential biomarker, demonstrating the ability to identify patients with CSP. Moreover, a combination of four lymphocyte parameters at baseline, relative lymphocyte counts, CD3+/CD69+, CD4+/CD45RO+, and CD4+/CD45RA+ab, was identified as a potential composite biomarker. This combination explains 23% of the variability in CSP, which is better than the best univariate parameter when compared to CD4+/CD45RO+ at baseline. Conclusions: The results suggest that other biomarkers can help monitor the conditions of patients and predict a favourable outcome. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

15 pages, 4006 KiB  
Article
IL-6, IL-12, and IL-23 STAT-Pathway Genetic Risk and Responsiveness of Lymphocytes in Patients with Multiple Sclerosis
by Marina R. von Essen, Helle B. Søndergaard, Eva R.S. Petersen and Finn Sellebjerg
Cells 2019, 8(3), 285; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8030285 - 26 Mar 2019
Cited by 21 | Viewed by 4384
Abstract
Multiple sclerosis (MS) is an immune-mediated demyelinating disease characterized by central nervous system (CNS) lymphocyte infiltration, abundant production of pro-inflammatory cytokines, and inappropriate activation of Th1 and Th17 cells, B cells, and innate immune cells. The etiology of MS is complex, and genetic [...] Read more.
Multiple sclerosis (MS) is an immune-mediated demyelinating disease characterized by central nervous system (CNS) lymphocyte infiltration, abundant production of pro-inflammatory cytokines, and inappropriate activation of Th1 and Th17 cells, B cells, and innate immune cells. The etiology of MS is complex, and genetic factors contribute to disease susceptibility. Genome-wide association studies (GWAS) have revealed numerous MS-risk alleles in the IL-6/STAT3, IL-12/STAT4, and IL-23/STAT3-pathways implicated in the differentiation of Th1 and Th17 cells. In this study, we investigated the signaling properties of these pathways in T, B, and NK cells from patients with relapsing-remitting MS (RRMS) and healthy controls, and assessed the genetic contribution to the activity of the pathways. This revealed a great variability in the level of STAT-pathway molecules and STAT activation between the cell types investigated. We also found a strong donor variation in IL-6, IL-12, and IL-23 responsiveness of primed CD4+ T cells. This variation could not be explained by a single MS-risk variant in a pathway component, or by an accumulation of multiple STAT-pathway MS-risk SNPs. The data of this study suggests that other factors in cohesion with the genetic background contribute to the responsiveness of the IL-6/STAT3, IL-12/STAT4, and IL-23/STAT3-pathways. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

14 pages, 1863 KiB  
Article
MiRNAs from DLK1-DIO3 Imprinted Locus at 14q32 are Associated with Multiple Sclerosis: Gender-Specific Expression and Regulation of Receptor Tyrosine Kinases Signaling
by Natalia Baulina, German Osmak, Ivan Kiselev, Ekaterina Popova, Alexey Boyko, Olga Kulakova and Olga Favorova
Cells 2019, 8(2), 133; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8020133 - 08 Feb 2019
Cited by 20 | Viewed by 4827
Abstract
Relapsing-remitting multiple sclerosis (RRMS) is the most prevalent course of multiple sclerosis. It is an autoimmune inflammatory disease of the central nervous system. To investigate the gender-specific involvement of microRNAs (miRNAs) in RRMS pathogenesis, we compared miRNA profiles in peripheral blood mononuclear cells [...] Read more.
Relapsing-remitting multiple sclerosis (RRMS) is the most prevalent course of multiple sclerosis. It is an autoimmune inflammatory disease of the central nervous system. To investigate the gender-specific involvement of microRNAs (miRNAs) in RRMS pathogenesis, we compared miRNA profiles in peripheral blood mononuclear cells separately in men and women (eight RRMS patients versus four healthy controls of each gender) using high-throughput sequencing. In contrast to women, six downregulated and 26 upregulated miRNAs (padj < 0.05) were identified in men with RRMS. Genes encoding upregulated miRNAs are co-localized in DLK1-DIO3 imprinted locus on human chromosome 14q32. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) analysis was performed in independent groups of men (16 RRMS patients and 10 healthy controls) and women (20 RRMS patients and 10 healthy controls). Increased expression of miR-431, miR-127-3p, miR-379, miR-376c, miR-381, miR-410 and miR-656 was again demonstrated in male (padj < 0.05), but not in female RRMS patients. At the same time, the expression levels of these miRNAs were lower in healthy men than in healthy women, whereas in RRMS men they increased and reached or exceeded levels in RRMS women. In general, we demonstrated that expression levels of these miRNAs depend both on “health–disease” status and gender. Network-based enrichment analysis identified that receptor tyrosine kinases-activated pathways were enriched with products of genes targeted by miRNAs from DLK1-DIO3 locus. These results suggest the male-specific involvement of these miRNAs in RRMS pathogenesis via regulation of PI3K/Akt signaling. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

23 pages, 6224 KiB  
Article
Expression of Translocator Protein and [18F]-GE180 Ligand Uptake in Multiple Sclerosis Animal Models
by Anne Nack, Matthias Brendel, Julia Nedelcu, Markus Daerr, Stella Nyamoya, Cordian Beyer, Carola Focke, Maximilian Deussing, Chloé Hoornaert, Peter Ponsaerts, Christoph Schmitz, Peter Bartenstein, Axel Rominger and Markus Kipp
Cells 2019, 8(2), 94; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8020094 - 28 Jan 2019
Cited by 32 | Viewed by 5143
Abstract
Positron emission tomography (PET) ligands targeting the translocator protein (TSPO) represent promising tools to visualize neuroinflammation in multiple sclerosis (MS). Although it is known that TSPO is expressed in the outer mitochondria membrane, its cellular localization in the central nervous system under physiological [...] Read more.
Positron emission tomography (PET) ligands targeting the translocator protein (TSPO) represent promising tools to visualize neuroinflammation in multiple sclerosis (MS). Although it is known that TSPO is expressed in the outer mitochondria membrane, its cellular localization in the central nervous system under physiological and pathological conditions is not entirely clear. The purpose of this study was to assess the feasibility of utilizing PET imaging with the TSPO tracer, [18F]-GE180, to detect histopathological changes during experimental demyelination, and to determine which cell types express TSPO. C57BL/6 mice were fed with cuprizone for up to 5 weeks to induce demyelination. Groups of mice were investigated by [18F]-GE180 PET imaging at week 5. Recruitment of peripheral immune cells was triggered by combining cuprizone intoxication with MOG35–55 immunization (i.e., Cup/EAE). Immunofluorescence double-labelling and transgene mice were used to determine which cell types express TSPO. [18F]-GE180-PET reliably detected the cuprizone-induced pathology in various white and grey matter regions, including the corpus callosum, cortex, hippocampus, thalamus and caudoputamen. Cuprizone-induced demyelination was paralleled by an increase in TSPO expression, glia activation and axonal injury. Most of the microglia and around one-third of the astrocytes expressed TSPO. TSPO expression induction was more severe in the white matter corpus callosum compared to the grey matter cortex. Although mitochondria accumulate at sites of focal axonal injury, these mitochondria do not express TSPO. In Cup/EAE mice, both microglia and recruited monocytes contribute to the TSPO expressing cell populations. These findings support the notion that TSPO is a valuable marker for the in vivo visualization and quantification of neuropathological changes in the MS brain. The pathological substrate of an increase in TSPO-ligand binding might be diverse including microglia activation, peripheral monocyte recruitment, or astrocytosis, but not axonal injury. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

18 pages, 3104 KiB  
Article
Biochemical Differences in Cerebrospinal Fluid between Secondary Progressive and Relapsing–Remitting Multiple Sclerosis
by Stephanie Herman, Torbjörn Åkerfeldt, Ola Spjuth, Joachim Burman and Kim Kultima
Cells 2019, 8(2), 84; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8020084 - 24 Jan 2019
Cited by 34 | Viewed by 6239
Abstract
To better understand the pathophysiological differences between secondary progressive multiple sclerosis (SPMS) and relapsing-remitting multiple sclerosis (RRMS), and to identify potential biomarkers of disease progression, we applied high-resolution mass spectrometry (HRMS) to investigate the metabolome of cerebrospinal fluid (CSF). The biochemical differences were [...] Read more.
To better understand the pathophysiological differences between secondary progressive multiple sclerosis (SPMS) and relapsing-remitting multiple sclerosis (RRMS), and to identify potential biomarkers of disease progression, we applied high-resolution mass spectrometry (HRMS) to investigate the metabolome of cerebrospinal fluid (CSF). The biochemical differences were determined using partial least squares discriminant analysis (PLS-DA) and connected to biochemical pathways as well as associated to clinical and radiological measures. Tryptophan metabolism was significantly altered, with perturbed levels of kynurenate, 5-hydroxytryptophan, 5-hydroxyindoleacetate, and N-acetylserotonin in SPMS patients compared with RRMS and controls. SPMS patients had altered kynurenine compared with RRMS patients, and altered indole-3-acetate compared with controls. Regarding the pyrimidine metabolism, SPMS patients had altered levels of uridine and deoxyuridine compared with RRMS and controls, and altered thymine and glutamine compared with RRMS patients. Metabolites from the pyrimidine metabolism were significantly associated with disability, disease activity and brain atrophy, making them of particular interest for understanding the disease mechanisms and as markers of disease progression. Overall, these findings are of importance for the characterization of the molecular pathogenesis of SPMS and support the hypothesis that the CSF metabolome may be used to explore changes that occur in the transition between the RRMS and SPMS pathologies. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

Review

Jump to: Research, Other

21 pages, 2338 KiB  
Review
The Adaptive Immune System in Multiple Sclerosis: An Estrogen-Mediated Point of View
by Alessandro Maglione, Simona Rolla, Stefania Federica De Mercanti, Santina Cutrupi and Marinella Clerico
Cells 2019, 8(10), 1280; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8101280 - 19 Oct 2019
Cited by 26 | Viewed by 4570
Abstract
Multiple sclerosis (MS) is a chronic central nervous system inflammatory disease that leads to demyelination and neurodegeneration. The third trimester of pregnancy, which is characterized by high levels of estrogens, has been shown to be associated with reduced relapse rates compared with the [...] Read more.
Multiple sclerosis (MS) is a chronic central nervous system inflammatory disease that leads to demyelination and neurodegeneration. The third trimester of pregnancy, which is characterized by high levels of estrogens, has been shown to be associated with reduced relapse rates compared with the rates before pregnancy. These effects could be related to the anti-inflammatory properties of estrogens, which orchestrate the reshuffling of the immune system toward immunotolerance to allow for fetal growth. The action of these hormones is mediated by the transcriptional regulation activity of estrogen receptors (ERs). Estrogen levels and ER expression define a specific balance of immune cell types. In this review, we explore the role of estradiol (E2) and ERs in the adaptive immune system, with a focus on estrogen-mediated cellular, molecular, and epigenetic mechanisms related to immune tolerance and neuroprotection in MS. The epigenome dynamics of immune systems are described as key molecular mechanisms that act on the regulation of immune cell identity. This is a completely unexplored field, suggesting a future path for more extensive research on estrogen-induced coregulatory complexes and molecular circuitry as targets for therapeutics in MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

14 pages, 322 KiB  
Review
The Molecular Basis for Remyelination Failure in Multiple Sclerosis
by Joel Gruchot, Vivien Weyers, Peter Göttle, Moritz Förster, Hans-Peter Hartung, Patrick Küry and David Kremer
Cells 2019, 8(8), 825; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8080825 - 03 Aug 2019
Cited by 67 | Viewed by 7294
Abstract
Myelin sheaths in the central nervous system (CNS) insulate axons and thereby allow saltatory nerve conduction, which is a prerequisite for complex brain function. Multiple sclerosis (MS), the most common inflammatory autoimmune disease of the CNS, leads to the destruction of myelin sheaths [...] Read more.
Myelin sheaths in the central nervous system (CNS) insulate axons and thereby allow saltatory nerve conduction, which is a prerequisite for complex brain function. Multiple sclerosis (MS), the most common inflammatory autoimmune disease of the CNS, leads to the destruction of myelin sheaths and the myelin-producing oligodendrocytes, thus leaving behind demyelinated axons prone to injury and degeneration. Clinically, this process manifests itself in significant neurological symptoms and disability. Resident oligodendroglial precursor cells (OPCs) and neural stem cells (NSCs) are present in the adult brain, and can differentiate into mature oligodendrocytes which then remyelinate the demyelinated axons. However, for multiple reasons, in MS the regenerative capacity of these cell populations diminishes significantly over time, ultimately leading to neurodegeneration, which currently remains untreatable. In addition, microglial cells, the resident innate immune cells of the CNS, can contribute further to inflammatory and degenerative axonal damage. Here, we review the molecular factors contributing to remyelination failure in MS by inhibiting OPC and NSC differentiation or modulating microglial behavior. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
29 pages, 625 KiB  
Review
Nanomodulation of Macrophages in Multiple Sclerosis
by Frances K. Nally, Chiara De Santi and Claire E. McCoy
Cells 2019, 8(6), 543; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8060543 - 05 Jun 2019
Cited by 47 | Viewed by 7057
Abstract
Multiple Sclerosis (MS) is a chronic demyelinating autoimmune disease primarily affecting young adults. Despite an unclear causal factor, symptoms and pathology arise from the infiltration of peripheral immune cells across the blood brain barrier. Accounting for the largest fraction of this infiltrate, macrophages [...] Read more.
Multiple Sclerosis (MS) is a chronic demyelinating autoimmune disease primarily affecting young adults. Despite an unclear causal factor, symptoms and pathology arise from the infiltration of peripheral immune cells across the blood brain barrier. Accounting for the largest fraction of this infiltrate, macrophages are functionally heterogeneous innate immune cells capable of adopting either a pro or an anti-inflammatory phenotype, a phenomenon dependent upon cytokine milieu in the CNS. This functional plasticity is of key relevance in MS, where the pro-inflammatory state dominates the early stage, instructing demyelination and axonal loss while the later anti-inflammatory state holds a key role in promoting tissue repair and regeneration in later remission. This review highlights a potential therapeutic benefit of modulating macrophage polarisation to harness the anti-inflammatory and reparative state in MS. Here, we outline the role of macrophages in MS and look at the role of current FDA approved therapeutics in macrophage polarisation. Moreover, we explore the potential of particulate carriers as a novel strategy to manipulate polarisation states in macrophages, whilst examining how optimising macrophage uptake via nanoparticle size and functionalisation could offer a novel therapeutic approach for MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

15 pages, 798 KiB  
Review
Pro-Thrombotic Activity of Blood Platelets in Multiple Sclerosis
by Joanna Saluk-Bijak, Angela Dziedzic and Michal Bijak
Cells 2019, 8(2), 110; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8020110 - 01 Feb 2019
Cited by 28 | Viewed by 5443
Abstract
The available data, including experimental studies, clearly indicate an excessive intravascular activation of circulating platelets in multiple sclerosis (MS) and their hyper-responsiveness to a variety of physiological activators. Platelet activation is manifested as an increased adhesion and aggregation and is accompanied by the [...] Read more.
The available data, including experimental studies, clearly indicate an excessive intravascular activation of circulating platelets in multiple sclerosis (MS) and their hyper-responsiveness to a variety of physiological activators. Platelet activation is manifested as an increased adhesion and aggregation and is accompanied by the formation of pro-thrombotic microparticles. Activated blood platelets also show an expression of specific membrane receptors, synthesis many of biomediators, and generation of reactive oxygen species. Epidemiological studies confirm the high risk of stroke or myocardial infarction in MS that are ischemic incidents, strictly associated with incorrect platelet functions and their over pro-thrombotic activity. Chronic inflammation and high activity of pro-oxidative processes in the course of MS are the main factors identified as the cause of excessive platelet activation. The primary biological function of platelets is to support vascular integrity, but the importance of platelets in inflammatory diseases is also well documented. The pro-thrombotic activity of platelets and their inflammatory properties play a part in the pathophysiology of MS. The analysis of platelet function capability in MS could provide useful information for studying the pathogenesis of this disease. Due to the complexity of pathological processes in MS, medication must be multifaceted and blood platelets can probably be identified as new targets for therapy in the future. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Graphical abstract

11 pages, 1657 KiB  
Review
Mechanism of Siponimod: Anti-Inflammatory and Neuroprotective Mode of Action
by Newshan Behrangi, Felix Fischbach and Markus Kipp
Cells 2019, 8(1), 24; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8010024 - 07 Jan 2019
Cited by 66 | Viewed by 9648
Abstract
Multiple sclerosis (MS) is a neuroinflammatory disorder of the central nervous system (CNS), and represents one of the main causes of disability in young adults. On the histopathological level, the disease is characterized by inflammatory demyelination and diffuse neurodegeneration. Although on the surface [...] Read more.
Multiple sclerosis (MS) is a neuroinflammatory disorder of the central nervous system (CNS), and represents one of the main causes of disability in young adults. On the histopathological level, the disease is characterized by inflammatory demyelination and diffuse neurodegeneration. Although on the surface the development of new inflammatory CNS lesions in MS may appear consistent with a primary recruitment of peripheral immune cells, questions have been raised as to whether lymphocyte and/or monocyte invasion into the brain are really at the root of inflammatory lesion development. In this review article, we discuss a less appreciated inflammation-neurodegeneration interplay, that is: Neurodegeneration can trigger the formation of new, focal inflammatory lesions. We summarize old and recent findings suggesting that new inflammatory lesions develop at sites of focal or diffuse degenerative processes within the CNS. Such a concept is discussed in the context of the EXPAND trial, showing that siponimod exerts anti-inflammatory and neuroprotective activities in secondary progressive MS patients. The verification or rejection of such a concept is vital for the development of new therapeutic strategies for progressive MS. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

22 pages, 2575 KiB  
Review
Can We Design a Nogo Receptor-Dependent Cellular Therapy to Target MS?
by Min Joung Kim, Jung Hee Kang, Paschalis Theotokis, Nikolaos Grigoriadis and Steven Petratos
Cells 2019, 8(1), 1; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8010001 - 20 Dec 2018
Cited by 19 | Viewed by 5803
Abstract
The current landscape of therapeutics designed to treat multiple sclerosis (MS) and its pathological sequelae is saturated with drugs that modify disease course and limit relapse rates. While these small molecules and biologicals are producing profound benefits to patients with reductions in annualized [...] Read more.
The current landscape of therapeutics designed to treat multiple sclerosis (MS) and its pathological sequelae is saturated with drugs that modify disease course and limit relapse rates. While these small molecules and biologicals are producing profound benefits to patients with reductions in annualized relapse rates, the repair or reversal of demyelinated lesions with or without axonal damage, remains the principle unmet need for progressive forms of the disease. Targeting the extracellular pathological milieu and the signaling mechanisms that drive neurodegeneration are potential means to achieve neuroprotection and/or repair in the central nervous system of progressive MS patients. The Nogo-A receptor-dependent signaling mechanism has raised considerable interest in neurological disease paradigms since it can promulgate axonal transport deficits, further demyelination, and extant axonal dystrophy, thereby limiting remyelination. If specific therapeutic regimes could be devised to directly clear the Nogo-A-enriched myelin debris in an expedited manner, it may provide the necessary CNS environment for neurorepair to become a clinical reality. The current review outlines novel means to achieve neurorepair with biologicals that may be directed to sites of active demyelination. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Graphical abstract

Other

Jump to: Research, Review

12 pages, 3281 KiB  
Brief Report
NK Cell Induced T Cell Anergy Depends on GRAIL Expression
by Grazyna Galazka, Malgorzata Domowicz, Alicja Ewiak-Paszynska and Anna Jurewicz
Cells 2019, 8(8), 790; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8080790 - 29 Jul 2019
Cited by 4 | Viewed by 3472
Abstract
NK cells (natural killer cells) being a part of the innate immune system have been shown to be involved in immunoregulation of autoimmune diseases. Previously we have shown that HINT1/Hsp70 treatment induced regulatory NK cells ameliorating experimental autoimmune encephalomyelitis (EAE) course and CD4+ [...] Read more.
NK cells (natural killer cells) being a part of the innate immune system have been shown to be involved in immunoregulation of autoimmune diseases. Previously we have shown that HINT1/Hsp70 treatment induced regulatory NK cells ameliorating experimental autoimmune encephalomyelitis (EAE) course and CD4+ T cells proliferation. NK cells were isolated from mice treated with HINT1/Hsp70 and co-cultured with proteolipid protein (PLP)-stimulated CD4+ T cells isolated from EAE mice. Cell proliferation was assessed by thymidine uptake, cytotoxicity by lactate dehydrogenase (LDH) release assay and fluorescence activated cell sorting (FACS) analysis, protein expression by Western blot, mRNA by quantitative RT-PCR. Gene related to anergy in lymphocytes (GRAIL) expression was downregulated by specific siRNA and GRAIL overexpression was induced by pcDNA-GRAIL transfection. HINT1/Hsp70 pretreatment of EAE SJL/J mice ameliorated EAE course, suppressed PLP-induced T cell proliferation by enhancing T cell expression of GRAIL as GRAIL downregulation restored T cell proliferation. HINT1/Hsp70 treatment induced immunoregulatory NK cells which inhibited PLP-stimulated T cell proliferation not depending on T cell necrosis and apoptosis. This immunoregulatory NK cell function depended on NK cell expression of GRAIL as GRAIL downregulation diminished inhibition of NK cell suppression of T cell proliferation. Similarly GRAIL overexpression in NK cells induced their regulatory function. HINT1/Hsp70 treatment generated regulatory NK cells characterized by expression of GRAIL. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Figure 1

6 pages, 1001 KiB  
Brief Report
Ocrelizumab Depletes CD20+ T Cells in Multiple Sclerosis Patients
by Stefan Gingele, Thais Langer Jacobus, Franz Felix Konen, Martin W. Hümmert, Kurt-Wolfram Sühs, Philipp Schwenkenbecher, Jonas Ahlbrecht, Nora Möhn, Lars H. Müschen, Lena Bönig, Sascha Alvermann, Reinhold E. Schmidt, Martin Stangel, Roland Jacobs and Thomas Skripuletz
Cells 2019, 8(1), 12; https://0-doi-org.brum.beds.ac.uk/10.3390/cells8010012 - 28 Dec 2018
Cited by 87 | Viewed by 16840
Abstract
Ocrelizumab, a humanized monoclonal anti-CD20 antibody, has shown pronounced effects in reduction of disease activity in multiple sclerosis (MS) patients and has recently been approved for the treatment of patients with relapsing MS (RMS) and primary progressive MS (PPMS). CD20 is mainly expressed [...] Read more.
Ocrelizumab, a humanized monoclonal anti-CD20 antibody, has shown pronounced effects in reduction of disease activity in multiple sclerosis (MS) patients and has recently been approved for the treatment of patients with relapsing MS (RMS) and primary progressive MS (PPMS). CD20 is mainly expressed by B cells, but a subset of T cells (CD3+CD20+ T cells) also expresses CD20, and these CD20+ T cells are known to be a highly activated cell population. The blood of MS patients was analyzed with multicolor flow cytometry before and two weeks after treatment with ocrelizumab regarding the phenotype of peripheral blood mononuclear cells. CD20-expressing CD3+ T cells were found in blood samples of all MS patients, accounted for 2.4% of CD45+ lymphocytes, and constituted a significant proportion (18.4%) of all CD20+ cells. CD3+CD20+ T cells and CD19+CD20+ B cells were effectively depleted two weeks after a single administration of 300 mg ocrelizumab. Our results demonstrate that treatment with ocrelizumab does not exclusively target B cells, but also CD20+ T cells, which account for a substantial amount of CD20-expressing cells. Thus, we speculate that the efficacy of ocrelizumab might also be mediated by the depletion of CD20-expressing T cells. Full article
(This article belongs to the Special Issue The Molecular and Cellular Basis for Multiple Sclerosis)
Show Figures

Graphical abstract

Back to TopTop