Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (36)

Search Parameters:
Keywords = Anti-MOG

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 3754 KiB  
Article
In Silico Modeling of Myelin Oligodendrocyte Glycoprotein Disulfide Bond Reduction by Phosphine-Borane Complexes
by Raheem Remtulla, Sanjoy Kumar Das and Leonard A. Levin
Pharmaceuticals 2024, 17(11), 1417; https://doi.org/10.3390/ph17111417 - 23 Oct 2024
Viewed by 486
Abstract
Background: Neurodegenerative diseases can cause vision loss by damaging retinal ganglion cells in the optic nerve. Novel phosphine-borane compounds (PBs) can protect these cells from oxidative stress via the reduction of disulfide bonds. However, the specific targets of these compounds are unknown. Proteomic [...] Read more.
Background: Neurodegenerative diseases can cause vision loss by damaging retinal ganglion cells in the optic nerve. Novel phosphine-borane compounds (PBs) can protect these cells from oxidative stress via the reduction of disulfide bonds. However, the specific targets of these compounds are unknown. Proteomic evidence suggests that myelin oligodendrocyte glycoprotein (MOG) is a potential target. MOG is of significant interest due to its role in anti-MOG optic neuritis syndrome. Methods: We used in silico modeling to explore the structural consequences of cleaving the extracellular domain MOG disulfide bond, both in isolation and in complex with anti-MOG antibodies. The potential binding of PBs to this bond was examined using molecular docking. Results: Cleaving the disulfide bond of MOG altered the structure of MOG dimers and reduced their energetic favorability by 46.13 kcal/mol. The energy profiles of anti-MOG antibody complexes were less favorable when the disulfide bond of MOG was reduced in the monomeric state by 55.21 kcal/mol, but the reverse was true in the dimeric state. PBs exhibited reducing capabilities with the MOG extracellular disulfide bond, with this best-scoring compound binding with an energy of −28.54 kcal/mol to the MOG monomer and −24.97 kcal/mol to the MOG dimer. Conclusions: These findings suggest that PBs can affect the structure of MOG dimers and the formation of antibody complexes by reducing the MOG disulfide bond. Structural changes in MOG could have implications for neurodegenerative diseases and anti-MOG syndrome. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Simplified schematic of MOG based on Kroepfl et al. proposed model [<a href="#B28-pharmaceuticals-17-01417" class="html-bibr">28</a>,<a href="#B29-pharmaceuticals-17-01417" class="html-bibr">29</a>].</p>
Full article ">Figure 2
<p>MOG dimer protein ribbon structure of Schrödinger protein–protein docking of extracellular MOG monomers with disulfide bond intact (PBD 1PKO).</p>
Full article ">Figure 3
<p>(<b>A</b>) MOG dimer protein ribbon structure of Schrödinger protein–protein docking of extracellular MOG monomers with disulfide bond cleaved pre dimer formation (PBD 1PKO). (<b>B</b>) Overlapping structures of MOG dimer protein ribbon structure with disulfide bond intact (RED) and MOG dimer protein ribbon structure with disulfide bond cleaved (YELLOW-GREEN) from Schrödinger protein–protein docking (<b>C</b>) Lateral view of overlapping structures of MOG dimer protein ribbon structure with disulfide bond intact (RED) and MOG dimer protein ribbon structure with disulfide bond cleaved (YELLOW-GREEN) from Schrödinger protein–protein docking. Measurement between arginine 126 on MOG dimer protein ribbon structure with disulfide bond intact and MOG dimer protein ribbon structure with disulfide bond cleaved in Ångstrom units.</p>
Full article ">Figure 4
<p>Protein ribbon structure of Schrödinger protein–protein docking-generated MOG dimer presented in the red structure (PBD 1PKO) with disulfide bond intact in complex with anti-MOG antibody presented in the grey structure(PBD 1PKQ). Two lateral orientations of the protein interaction are depicted.</p>
Full article ">Figure 5
<p>Protein ribbon structure of Schrödinger protein–protein docking-generated MOG dimer presented in the yellow-green structure (PBD 1PKO) with disulfide bond cleaved pre dimer formation in complex with anti-MOG antibody presented in the grey structure (PBD 1PKQ). Two lateral orientations of the protein interaction are depicted.</p>
Full article ">Figure 6
<p>Overlapping structures of MOG dimer with disulfide bond intact presented in the red structure in complex with anti-MOG antibody presented in the grey structure (PBD 1PKQ), and MOG dimer with disulfide bond cleaved pre dimer formation presented in the yellow-green structure in complex with anti-MOG antibody presented in the grey structure (PBD 1PKQ). Two lateral orientations of the protein interaction are depicted.</p>
Full article ">Figure 7
<p>(<b>A</b>) Ligand interaction diagram of covalent docking of triphenylphosphine to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>B</b>) Ligand interaction diagram of covalent docking of tris(2-carboxyethyl)phosphine to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>C</b>) Ligand interaction diagram of covalent docking of P1 to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>D</b>) Ligand interaction diagram of covalent docking of P1 monoacid R to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>E</b>) Ligand interaction diagram of covalent docking of P1-monoacid S to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>F</b>) Ligand interaction diagram of covalent docking of P1-diacid to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>G</b>) Ligand interaction diagram of covalent docking of P2 to cystine 24 on myelin oligodendrocyte glycoprotein. (<b>H</b>) Ligand interaction diagram of covalent docking of P2 acid to cystine 24 on myelin oligodendrocyte glycoprotein.</p>
Full article ">Figure 8
<p>(<b>a</b>) Covalent docking results of P1-diacid (ball and stick) in complex with cystine 24 on ribbon diagram of MOG monomer (PBD 1PKO). (<b>b</b>) Binding site map on MOG monomer (PBD 1PKO) overlayed with covalent docking results of P1-diacid (ball and stick) in complex with cystine 24 on ribbon diagram of MOG monomer (PBD 1PKO).</p>
Full article ">Figure 9
<p>Phosphine-borane structures. Chemical structures of (<b>a</b>) PB1, (<b>b</b>) PB2 (<b>c</b>) PB-Ph, and (<b>d</b>) PB-Me.</p>
Full article ">
20 pages, 5292 KiB  
Article
Cellular and Immunological Analysis of 2D2/Th Hybrid Mice Prone to Experimental Autoimmune Encephalomyelitis in Comparison with 2D2 and Th Lines
by Kseniya S. Aulova, Andrey E. Urusov, Aleksander D. Chernyak, Ludmila B. Toporkova, Galina S. Chicherina, Valentina N. Buneva, Irina A. Orlovskaya and Georgy A. Nevinsky
Int. J. Mol. Sci. 2024, 25(18), 9900; https://doi.org/10.3390/ijms25189900 - 13 Sep 2024
Viewed by 591
Abstract
Previously, we described the mechanisms of development of autoimmune encephalomyelitis (EAE) in 3-month-old C57BL/6, Th, and 2D2 mice. The faster and more profound spontaneous development of EAE with the achievement of deeper pathology occurs in hybrid 2D2/Th mice. Here, the cellular and immunological [...] Read more.
Previously, we described the mechanisms of development of autoimmune encephalomyelitis (EAE) in 3-month-old C57BL/6, Th, and 2D2 mice. The faster and more profound spontaneous development of EAE with the achievement of deeper pathology occurs in hybrid 2D2/Th mice. Here, the cellular and immunological analysis of EAE development in 2D2/Th mice was carried out. In Th, 2D2, and 2D2/Th mice, the development of EAE is associated with a change in the differentiation profile of hemopoietic bone marrow stem cells, which, in 2D2/Th, differs significantly from 2D2 and Th mice. Hybrid 2D2/Th mice demonstrate a significant difference in these changes in all strains of mice, leading to the production of antibodies with catalytic activities, known as abzymes, against self-antigens: myelin oligodendrocyte glycoprotein (MOG), DNA, myelin basic protein (MBP), and five histones (H1–H4) hydrolyze these antigens. There is also the proliferation of B and T lymphocytes in different organs (blood, bone marrow, thymus, spleen, lymph nodes). The patterns of changes in the concentration of antibodies and the relative activity of abzymes during the spontaneous development of EAE in the hydrolysis of these immunogens are significantly or radically different for the three lines of mice: Th, 2D2, and 2D2/Th. Several factors may play an essential role in the acceleration of EAE in 2D2/Th mice. The treatment of mice with MOG accelerates the development of EAE pathology. In the initial period of EAE development, the concentration of anti-MOG antibodies in 2D2/Th is significantly higher than in Th (29.1-fold) and 2D2 (11.7-fold). As shown earlier, antibodies with DNase activity penetrate cellular and nuclear membranes and activate cell apoptosis, stimulating autoimmune processes. In the initial period of EAE development, the concentration of anti-DNA antibodies in 2D2/Th hybrids is higher than in Th (4.6-fold) and 2D2 (25.7-fold); only 2D2/Th mice exhibited a very strong 10.6-fold increase in the DNase activity of IgGs during the development of EAE. Free histones in the blood are cytotoxic and stimulate the development of autoimmune diseases. Only in 2D2/Th mice, during different periods of EAE development, was a sharp increase in the anti-antibody activity in the hydrolysis of some histones observed. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

Figure 1
<p>Comparison of proteinuria levels in 3-month-old C57BL/6, 2D2, Th, and hybrid 2D2/Th mice. For comparison, data for 2D2 and Th mice are taken from the works [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>]. The difference between the data of all curves is characterized by <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Changes in BFU-E (<b>A</b>), CFU-E (<b>B</b>), CFU-GM (<b>C</b>), and CFU-GEMM (<b>D</b>) cell colony units during development of EAE in C57BL/6, Th, 2D2, and hybrid 2D2/Th mice. The relative number of all colonies was calculated for 15,000 bone marrow cells. For comparison, data for C57BL/6, 2D2, and Th mice are taken from the works [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>]. The difference between the data of all curves is characterized by <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Changes in the content of B cells in different organs of Th, 2D2, and 2D2/Th mice: blood (<b>A</b>), bone marrow (<b>B</b>), thymus (<b>C</b>), spleen (<b>D</b>), and lymph nodes (<b>E</b>). Data for 2D2 and Th mice are taken from the works [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>] for comparison. The difference between the data of all curves is characterized by <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Changes in the T cell content in different organs of Th, 2D2, and 2D2/Th mice: blood (<b>A</b>), bone marrow (<b>B</b>), thymus (<b>C</b>), spleen (<b>D</b>), and lymph nodes (<b>E</b>). For comparison, data for 2D2 and Th mice are taken from the works [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>]. The difference between the data of all curves is characterized by <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>The changes in the concentration of antibodies against DNA (<b>A</b>), MBP (<b>B</b>), MOG (<b>C</b>), and five histones (<b>D</b>) during development of EAE in C57BL/6, Th, 2D2, and hybrid 2D2/Th mice. Dependencies corresponding to different lines of mice are marked in the panels. For comparison, data for C57BL/6, 2D2, and Th mice are taken from the works [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>]. The difference between the data of all curves is characterized by <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>The IgG<sub>mix</sub> (mixture of 21 IgG preparations; 17 μg) homogeneity analysis by SDS-PAGE with Coomassie staining (<b>A</b>). Panel (<b>A</b>) demonstrates the position of IgG<sub>mix</sub>. The relative activities in the hydrolysis of DNA (•), MOG (□), MBP (<span style="color:red">★</span>), and histones (∆) were determined using eluates of many gel fragments (2–3 mm) (<b>B</b>). After substrate incubation for 24 h with the eluates from gel, complete hydrolysis of four antigens was taken for 100% (<b>B</b>). The errors of the activities from 2 independent experiments did not exceed 7–10%.</p>
Full article ">Figure 7
<p>Changes over time in DNase activity of IgG-abzymes (<b>A</b>), as well as MBP- (<b>B</b>) and MOG-hydrolyzing activities (<b>C</b>). Dependencies corresponding to hybrid 2D2/Th, 2D2, Th, and C57BL/6 mice are given in different colors. The data for Th, 2D2, and C57BL/6 mice are given for comparison from our previously published articles [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>].</p>
Full article ">Figure 8
<p>Changes over time in protease activities of IgG-abzymes hydrolyzing H1 (<b>A</b>), H2A (<b>B</b>), H2B (<b>C</b>), H3 (<b>D</b>), and H4 (<b>E</b>) histones. Dependencies corresponding to hybrid 2D2/Th, 2D2, Th, and C57BL/6 mice are given in different colors. The data for Th, 2D2, and C57BL/6 mice are given for comparison from our previously published articles [<a href="#B29-ijms-25-09900" class="html-bibr">29</a>,<a href="#B30-ijms-25-09900" class="html-bibr">30</a>,<a href="#B31-ijms-25-09900" class="html-bibr">31</a>,<a href="#B32-ijms-25-09900" class="html-bibr">32</a>]. The difference between the data of all curves is characterized by <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>The comparison of relative DNase (<b>A</b>), MOG- (<b>B</b>), and MBP-hydrolyzing (<b>C</b>) activities of 3-month-old 2D2/Th paralyzed mice with non-paralyzed 2D2/Th, 2D2, and Th mice. The differences between the data of all curves (<span class="html-italic">p</span>) are given in the terx.</p>
Full article ">
10 pages, 1602 KiB  
Case Report
Isolated Intracranial Hypertensions as Onset of Myelin Oligodendrocyte Glycoprotein Antibody Disease
by Laura Papetti, Giulia Moltoni, Daniela Longo, Gabriele Monte, Francesco Dellepiane, Stefano Pro, Giorgia Bracaglia, Claudia Ruscitto, Alberto Verrotti and Massimiliano Valeriani
J. Clin. Med. 2024, 13(15), 4468; https://doi.org/10.3390/jcm13154468 - 30 Jul 2024
Cited by 1 | Viewed by 1161
Abstract
Myelin oligodendrocyte glycoprotein antibody disease (MOGAD) is characterized by multiple phenotypic conditions such as acute disseminated encephalomyelitis, optic neuritis, and myelitis. MOGAD’s spectrum is expanding, with potential symptoms of increased intracranial pressure that are similar to idiopathic intracranial hypertension (IIH). We report a [...] Read more.
Myelin oligodendrocyte glycoprotein antibody disease (MOGAD) is characterized by multiple phenotypic conditions such as acute disseminated encephalomyelitis, optic neuritis, and myelitis. MOGAD’s spectrum is expanding, with potential symptoms of increased intracranial pressure that are similar to idiopathic intracranial hypertension (IIH). We report a boy with new-onset continuous headache and a brain MRI at onset suggesting idiopathic intracranial hypertension (IIH). The patient showed resistance to treatment with acetazolamide and, after one month, developed optic neuritis in the left eye. Laboratory tests documented positive MOG antibodies (anti-MOG) in the serum. The final diagnosis was MOGAD, with the initial symptoms resembling IIH. Full article
(This article belongs to the Special Issue Advances in Child Neurology)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Axial T2-weighted CISS (Constructive Interference Steady State); (<b>b</b>) sagittal T2-weighted CISS; (<b>c</b>) axial FLAIR (Fluid Attenuated Inversion Recovery; (<b>d</b>) sagittal T2 FLAIR; (<b>e</b>) sagittal subtraction images between post-contrast and pre-contrast T1 weighted MPRAGE. (<b>a</b>,<b>b</b>) Finding consistent with idiomatic intracranic hypertension (IIH): bilateral prominent subarachnoid space around the optic nerves (arrow), more evident on the right side because of the left optic nerve swelling, intraocular protrusion of the optic nerves (arrowhead) (<b>a</b>), and partially empty sella (arrow) (<b>b</b>); (<b>c</b>–<b>e</b>) findings consistent with optic neuritis (ON): T2-FLAIR hyperintensity and swelling of the left optic nerve (<b>c</b>,<b>d</b>) associated with contrast enhancement (<b>e</b>).</p>
Full article ">Figure 2
<p><b>Visual Evoked Potentials.</b> Black and white pattern reversal visual evoked potentials (PR-VEP) with a visual angle of 15 min of arc were recorded separately for each eye through silver–silver chloride cup electrodes placed over Oz and Cz (reference) with ground on the ear lobe. P100 latencies are reported for each PR-VEP. PR-VEPs performed at early onset of disease were bilaterally normal (t0). After one week (t1), P100 was absent on the left eye, but was still normal on the right eye. During follow-up, PR-VEPs showed gradual improvement on the left eye after eight days (t2), about one month (t3), and three months (t4) from the onset of disease. Finally a slight asymmetry of P100 latency was observed, increased by 10 ms on the left eye compared to the contralateral eye.</p>
Full article ">
13 pages, 1644 KiB  
Article
IL-6 Inhibition as a Therapeutic Target in Aged Experimental Autoimmune Encephalomyelitis
by María Dema, Herena Eixarch, Mireia Castillo, Xavier Montalban and Carmen Espejo
Int. J. Mol. Sci. 2024, 25(12), 6732; https://doi.org/10.3390/ijms25126732 - 19 Jun 2024
Viewed by 1235
Abstract
Multiple sclerosis (MS) onset at an advanced age is associated with a higher risk of developing progressive forms and a greater accumulation of disability for which there are currently no effective disease-modifying treatments. Immunosenescence is associated with the production of the senescence-associated secretory [...] Read more.
Multiple sclerosis (MS) onset at an advanced age is associated with a higher risk of developing progressive forms and a greater accumulation of disability for which there are currently no effective disease-modifying treatments. Immunosenescence is associated with the production of the senescence-associated secretory phenotype (SASP), with IL-6 being one of the most prominent cytokines. IL-6 is a determinant for the development of autoimmunity and neuroinflammation and is involved in the pathogenesis of MS. Herein, we aimed to preclinically test the therapeutic inhibition of IL-6 signaling in experimental autoimmune encephalomyelitis (EAE) as a potential age-specific treatment for elderly MS patients. Young and aged mice were immunized with myelin oligodendrocyte protein (MOG)35–55 and examined daily for neurological signs. Mice were randomized and treated with anti-IL-6 antibody. Inflammatory infiltration was evaluated in the spinal cord and the peripheral immune response was studied. The blockade of IL-6 signaling did not improve the clinical course of EAE in an aging context. However, IL-6 inhibition was associated with an increase in the peripheral immunosuppressive response as follows: a higher frequency of CD4 T cells producing IL-10, and increased frequency of inhibitory immune check points PD-1 and Tim-3 on CD4+ T cells and Lag-3 and Tim-3 on CD8+ T cells. Our results open the window to further studies aimed to adjust the anti-IL-6 treatment conditions to tailor an effective age-specific therapy for elderly MS patients. Full article
(This article belongs to the Special Issue Multiple Sclerosis and the Neuroinflammation)
Show Figures

Figure 1

Figure 1
<p>Treatment with anti-IL-6 did not ameliorate the clinical course nor reduce CNS inflammatory infiltration in aged EAE. (<b>A</b>) Clinical course, (<b>B</b>) weight loss, (<b>C</b>) time to reach a score of 3 (mild tetraparesis) and (<b>D</b>) time to reach a score of 4 (tetraparesis) in young and aged mice treated with anti-IL-6 antibody or IgG isotype. Data represent two pooled independent experiments with nine mice per treatment and age for each experiment, accounting for a total of n = 18 (IgG isotype) and n = 18 (anti-IL-6 antibody) for young mice, and n = 18 (IgG isotype) and n = 18 (anti-IL-6 antibody) for aged mice. Dotted lines indicate the treatment initiation period. (<b>E</b>) Quantification of inflammatory infiltration in the white matter (WM) spinal cord in young and aged mice treated with Ig isotype or anti-IL-6 antibody. Data represent an individual experiment with n = 9 (IgG isotype) and n = 8 (anti-IL-6 antibody) for young mice and n = 9 (IgG isotype) and n = 7 (anti-IL-6 antibody) for aged mice. Only mice that reached the endpoint were included in the analysis. Variables were analyzed using a two-way ANOVA test and statistical significance correction for multiple comparisons was performed with Bonferroni adjustment in (<b>A</b>,<b>B</b>), Log-rank test in (<b>C</b>,<b>D</b>) and <span class="html-italic">t</span>-test in (<b>E</b>). Data are expressed as the mean ± SEM in (<b>A</b>,<b>B</b>), as survival curves in (<b>C</b>,<b>D</b>) and as mean ± SD in (<b>E</b>). AUC: area under the curve; H&amp;E: hematoxylin and eosin; WM: white matter.</p>
Full article ">Figure 2
<p>Anti-IL-6 antibody treatment increased peripheral immunosuppressive response in aged EAE. (<b>a</b>) Th1/Th17 ratio, (<b>b</b>) Th1/Treg ratio, (<b>c</b>) Th17/Treg ratio, (<b>d</b>) Th1/Th2 ratio, (<b>e</b>) PD-1<sup>+</sup>CD4<sup>+</sup>, (<b>f</b>) LAG-3<sup>+</sup>CD4<sup>+</sup>, (<b>g</b>) TIM-3<sup>+</sup>CD4<sup>+</sup>, (<b>h</b>) PD-1<sup>+</sup>CD8<sup>+</sup>, (<b>i</b>) LAG-3<sup>+</sup>CD8<sup>+</sup>, (<b>j</b>) TIM-3<sup>+</sup>CD8<sup>+</sup> T effector cells at 28 days postimmunization in young and aged mice treated with IgG isotype or antiIL-6 antibody. Data represent two pooled independent experiments with eight to nine mice per treatment and age for each experiment, accounting for a total of n = 18 (IgG isotype) and n = 18 (anti-IL-6 antibody) for young mice and n = 17 (IgG isotype) and n = 17 (anti-IL-6 antibody) for aged mice. Only mice that reached the endpoint were included in the analysis. Variables were analyzed using a GLIMMIX test and statistical significance correction for multiple comparisons was performed with Bonferroni adjustment. Data are expressed as the ratio of cell frequencies in (<b>a</b>–<b>d</b>) and as mean ± SD in (<b>e</b>–<b>j</b>). * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01. Te: effector T; Th: helper T; Treg: regulatory T.</p>
Full article ">Figure 3
<p>Scheme of the immunization conditions and experimental design of this study. Created with BioRender.com, accessed on 10 June 2024.</p>
Full article ">
17 pages, 4595 KiB  
Article
Digital Pathology Identifies Associations between Tissue Inflammatory Biomarkers and Multiple Sclerosis Outcomes
by Benjamin Cooze, James Neal, Alka Vineed, J. C. Oliveira, Lauren Griffiths, K. H. Allen, Kristen Hawkins, Htoo Yadanar, Krisjanis Gerhards, Ildiko Farkas, Richard Reynolds and Owain Howell
Cells 2024, 13(12), 1020; https://doi.org/10.3390/cells13121020 - 11 Jun 2024
Viewed by 1208
Abstract
Background: Multiple sclerosis (MS) is a clinically heterogeneous disease underpinned by inflammatory, demyelinating and neurodegenerative processes, the extent of which varies between individuals and over the course of the disease. Recognising the clinicopathological features that most strongly associate with disease outcomes will inform [...] Read more.
Background: Multiple sclerosis (MS) is a clinically heterogeneous disease underpinned by inflammatory, demyelinating and neurodegenerative processes, the extent of which varies between individuals and over the course of the disease. Recognising the clinicopathological features that most strongly associate with disease outcomes will inform future efforts at patient phenotyping. Aims: We used a digital pathology workflow, involving high-resolution image acquisition of immunostained slides and opensource software for quantification, to investigate the relationship between clinical and neuropathological features in an autopsy cohort of progressive MS. Methods: Sequential sections of frontal, cingulate and occipital cortex, thalamus, brain stem (pons) and cerebellum including dentate nucleus (n = 35 progressive MS, females = 28, males = 7; age died = 53.5 years; range 38–98 years) were immunostained for myelin (anti-MOG), neurons (anti-HuC/D) and microglia/macrophages (anti-HLA). The extent of demyelination, neurodegeneration, the presence of active and/or chronic active lesions and quantification of brain and leptomeningeal inflammation was captured by digital pathology. Results: Digital analysis of tissue sections revealed the variable extent of pathology that characterises progressive MS. Microglia/macrophage activation, if found at a higher level in a single block, was typically elevated across all sampled blocks. Compartmentalised (perivascular/leptomeningeal) inflammation was associated with age-related measures of disease severity and an earlier death. Conclusion: Digital pathology identified prognostically important clinicopathological correlations in MS. This methodology can be used to prioritise the principal pathological processes that need to be captured by future MS biomarkers. Full article
(This article belongs to the Special Issue New Advances in Neuroinflammation)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Systematic sampling of tissue blocks for immunostaining and quantification by digital pathology (<b>A</b>). Postmortem human brain tissue was sampled (<b>left</b> to <b>right</b>) at the level of the pons, cerebellum (including dentate nucleus; CbDN), occipital pole (Occ), superior frontal gyrus (SFG), cingulate gyrus (CG) and thalamus (Thal). Created with BioRender.com. Sections were prepared for immunohistochemistry, slide digitisation and quantitative pathology ((<b>B</b>); anti-MOG immunostaining to reveal areas of demyelination—yellow annotations), neuron density ((<b>C</b>); anti-HUC and red colour mask depicting automatically detected HUC+ neurons) and microglia/macrophages ((<b>D</b>); anti-HLA and red colour mask). Scale bars; (<b>B</b>) = 2 mm; (<b>C</b>,<b>D</b>) = 50 µm.</p>
Full article ">Figure 2
<p>Quantifying inflammation, demyelination and neurodegeneration to reveal widespread HLA+ microglia/macrophage activation in progressive MS. Each case was further classified according to the presence/absence of active or chronic active demyelination (red image outline; (<b>A</b>)) and the relative extent of perivascular/leptomeningeal inflammation (LFB/CFV examples of infiltrates graded 1/3 and 3/3, respectively, MS330). Forty percent of cases harboured ≥1 active or chronic active lesion (<b>B</b>). Moderate (grade 2/3) or substantial (grade 3/3) levels of cellular infiltrates were noted in 45% of cases (<b>C</b>). Scatter plots representing average area of HLA + immunoreactivity (<b>D</b>), neuron density ((<b>E</b>); HUC+) and lesion area ((<b>F</b>); MOG) per region of interest, per case (group mean and standard deviation). Heat maps representing associations between microglia/macrophage density (<b>G</b>), neuron density (<b>H</b>) and lesion area (<b>I</b>) across the sampled brain regions (Spearman r values quoted). HLA+ microglia/macrophage activation in one block tended to be reflected by an elevated signal in other blocks from the same case ((<b>G</b>,<b>J</b>–<b>L</b>); representative images of SFG, CG and Thal blocks from a case with a low level of diffuse HLA+ immunosignal in comparison to a case with widespread microglia/macrophage activation; ‘HLA- high’). Images of HLA+ microglia (<b>K</b>,<b>L</b>) captured from the areas represented in the thalamus block. Significant correlations and exact p values are described in the result text. Images of HLA+ microglia (<b>K</b>,<b>L</b>)captured from areas represented in the thalamus block by dashed boxes. Abbreviations: C active, chronic active demyelinating lesion; SFG, superior frontal gyrus; CG, cingulate gyrus; Thal, thalamus; Occ, occipital pole; Cb DN, cerebellar white matter and dentate nucleus. Scale bars: (<b>A</b>) = 100 µm; (<b>J</b>) = 4 mm; (<b>K</b>,<b>L</b>) = 20 µm.</p>
Full article ">Figure 3
<p>Comparing pathological variables and pathological–clinical associations. Spearman correlation analysis comparing microglia/macrophage activation in the normal appearing tissue, presence of active demyelination (Active/SEL: yes) or extent of perivascular/meningeal inflammation (Men/PV infilt; (<b>A</b>)) with neuron density (HUC) and lesion area (MOG) across the six sampled brain blocks per case. Comparing microglia/macrophage activation, presence of active lesions (Active/SEL: yes) and the relative extent of perivascular/leptomeningeal inflammation (Men/PV infilt) with clinical measures of disease severity (<b>B</b>). Note the presence of active lesions and the relative extent of perivascular/leptomeningeal inflammation with age at MS onset (Age_onset), age at secondary progression (Age_prog), age at wheelchair (Age_WC) and age died. Scale: White indicates no correlation; blue indicates positive associations; and red indicates negative associations. Individual Spearman r values are quoted for each comparison and significant correlations and exact p values are described in the results text. Abbreviations: SFG, superior frontal gyrus; CG, cingulate gyrus; Thal, thalamus; Occ, occipital pole; Cb DN, cerebellar white matter and dentate nucleus.</p>
Full article ">Figure 4
<p>Phenotyping on the basis of the measured extent of inflammation reveals MS groups that differ by clinical outcomes. Box and whisker plots representing average percent area of HLA+ stain (<b>A</b>), neuron density (<b>B</b>) and lesion area (<b>C</b>) per case, grouped according to their relative extent of perivascular/leptomeningeal inflammation (PVI/Men rated 0–1 versus PVI/Men rated 2–3). Median, inter-quartile range and min–max values are represented. Data compared by non-parametric T-test. Cases grouped according to their relative extent of perivascular/leptomeningeal inflammation differed significantly in terms of the finding of active lesions ((<b>D</b>); Fisher’s exact test) and age at key clinical milestones ((<b>E</b>–<b>H</b>); PVI/Men rated 0–1, green data points; PVI/Men rated 2–3, red data points). Perivascular/leptomeningeal inflammation (<b>I</b>) was associated with increased density of CD3+ and CD20+ immunostaining in demyelinated thalamus, occipital pole white matter (Occ WM) and grey matter (Occ GM) in comparison to matched normal-appearing tissue (<b>J</b>). Increased CD3 and CD20+ staining associated with a younger age of death (Age died), a shorter interval to secondary progression (Onset to prog) and a shorter overall disease length (Onset to death). Spearman r values included in the heat map (<b>K</b>) and exact p values are included in the result text. Abbreviations: PVI/Men rated 0–1 or 2–3, cases grouped according to relative extent of perivascular/leptomeningeal inflammation.</p>
Full article ">
17 pages, 2257 KiB  
Article
Clinical Research into Central Nervous System Inflammatory Demyelinating Diseases Related to COVID-19 Vaccines
by Mei-Yun Cheng, Hsuan-Chen Ho, Jung-Lung Hsu, Yi Wang, Linyi Chen, Siew-Na Lim, Ming-Feng Liao and Long-Sun Ro
Diseases 2024, 12(3), 60; https://doi.org/10.3390/diseases12030060 - 20 Mar 2024
Viewed by 3114
Abstract
Various vaccines have been developed in response to the SARS-CoV-2 pandemic, and the safety of vaccines has become an important issue. COVID-19 vaccine-related central nervous system inflammatory demyelinating diseases (CNS IDDs) have been reported recently. We present one case of AstraZeneca vaccine-related myelin [...] Read more.
Various vaccines have been developed in response to the SARS-CoV-2 pandemic, and the safety of vaccines has become an important issue. COVID-19 vaccine-related central nervous system inflammatory demyelinating diseases (CNS IDDs) have been reported recently. We present one case of AstraZeneca vaccine-related myelin oligodendrocyte glycoprotein (MOG) antibody-associated disease and a literature review of another 78 patients published from January 2020 to October 2022. Patients were divided into three vaccine types (viral vector, mRNA, and inactivated vaccines) for further analyses. Among 79 patients with COVID-19 vaccine-related CNS IDDs, 49 (62%) cases received viral vector vaccines, 20 (25.3%) received mRNA vaccines, and 10 (12.7%) received inactivated vaccines. Twenty-seven cases (34.2%) were confirmed with autoantibodies, including fifteen patients (19%) with anti-MOG, eleven (13.9%) with anti-aquaporin 4 (AQP4), and one (1.3%) with both antibodies. Significantly, more males developed CNS IDDs post viral vector vaccines compared to mRNA and inactivated vaccines. Patients receiving mRNA vaccines were older than those receiving other types. Furthermore, mRNA and inactivated vaccines correlated more with anti-AQP4 antibodies, while viral vector vaccines showed higher MOG positivity. This research suggests potential associations between COVID-19 vaccine-related CNS IDDs and gender, age, and autoantibodies, contingent on vaccine types. Protein sequence analysis implies similarities between the S protein and AQP4/MOG. Further studies may elucidate the mechanisms of CNS IDDs, aiding vaccine selection for specific types. Full article
Show Figures

Figure 1

Figure 1
<p>Spinal MRI: On the 6th day after onset, an intramedullary spotty lesion with T2 hyperintensity at T3 to T4 was shown (red arrows: (<b>a</b>) sagittal view. (<b>c</b>) axial view). Five months later, the lesion over T3 to T4 was totally resolved on T2 FLAIR images (red arrows: (<b>b</b>) sagittal view. (<b>d</b>) axial view).</p>
Full article ">Figure 2
<p>Brain MRI: Bilateral poorly demarcated T2 FLAIR hyperintensities at brainstem (<b>a</b>–<b>c</b>), pulvinar thalami (<b>d</b>), putamen (<b>d</b>), centrum semiovale (<b>e</b>), and subcortical white matters (<b>f</b>) are marked by red arrows in the image.</p>
Full article ">Figure 3
<p>Cell-based immunofluorescence assay (Euroimmun, Lübeck, Germany): Indirect immunofluorescence test was used on HEK-293 cells transfected with plasmids containing MOG. Compared to the negative control (<b>a</b>), positive fluorescence was observed over the cell membrane and cytoplasm in transfected HEK-293 cells after applying the diluted (1:10) patient’s serum (<b>b</b>) and FITC-labeled secondary anti-human IgG antibody at 20× magnification.</p>
Full article ">Figure 4
<p>S-Covid 19 refers to the spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), retrieved from the Protein Data Bank (PDB) under the accession number 6X29. Aquaporin 4 (AQP4) has a well-documented 3D structure with established structural domains, sourced from the PDB with the accession number 3GD8 (<b>a</b>). Notably, the extracellular loop of the AQP4 transmembrane protein can function as an antigen or be recognized by antibodies. The 3D structure of MOG is unknown (<b>b</b>). The Expasy SIM protein sequence alignment tool was utilized for analysis (<a href="https://web.expasy.org/sim/" target="_blank">https://web.expasy.org/sim/</a> accessed on 6 June 2023). Additionally, we employed PyMOL for protein structural analysis. The red-highlighted structure represents regions of high similarity between the two proteins.</p>
Full article ">
11 pages, 429 KiB  
Review
Pathogenesis, Clinical Features, and Treatment of Patients with Myelin Oligodendrocyte Glycoprotein (MOG) Autoantibody-Associated Disorders Focusing on Optic Neuritis with Consideration of Autoantibody-Binding Sites: A Review
by Keiko Tanaka, Takeshi Kezuka, Hitoshi Ishikawa, Masami Tanaka, Kenji Sakimura, Manabu Abe and Meiko Kawamura
Int. J. Mol. Sci. 2023, 24(17), 13368; https://doi.org/10.3390/ijms241713368 - 29 Aug 2023
Cited by 5 | Viewed by 2406
Abstract
Although there is a substantial amount of data on the clinical characteristics, diagnostic criteria, and pathogenesis of myelin oligodendrocyte glycoprotein (MOG) autoantibody-associated disease (MOGAD), there is still uncertainty regarding the MOG protein function and the pathogenicity of anti-MOG autoantibodies in this disease. It [...] Read more.
Although there is a substantial amount of data on the clinical characteristics, diagnostic criteria, and pathogenesis of myelin oligodendrocyte glycoprotein (MOG) autoantibody-associated disease (MOGAD), there is still uncertainty regarding the MOG protein function and the pathogenicity of anti-MOG autoantibodies in this disease. It is important to note that the disease characteristics, immunopathology, and treatment response of MOGAD patients differ from those of anti-aquaporin 4 antibody-positive neuromyelitis optica spectrum disorders (NMOSDs) and multiple sclerosis (MS). The clinical phenotypes of MOGAD are varied and can include acute disseminated encephalomyelitis, transverse myelitis, cerebral cortical encephalitis, brainstem or cerebellar symptoms, and optic neuritis. The frequency of optic neuritis suggests that the optic nerve is the most vulnerable lesion in MOGAD. During the acute stage, the optic nerve shows significant swelling with severe visual symptoms, and an MRI of the optic nerve and brain lesion tends to show an edematous appearance. These features can be alleviated with early extensive immune therapy, which may suggest that the initial attack of anti-MOG autoantibodies could target the structures on the blood–brain barrier or vessel membrane before reaching MOG protein on myelin or oligodendrocytes. To understand the pathogenesis of MOGAD, proper animal models are crucial. However, anti-MOG autoantibodies isolated from patients with MOGAD do not recognize mouse MOG efficiently. Several studies have identified two MOG epitopes that exhibit strong affinity with human anti-MOG autoantibodies, particularly those isolated from patients with the optic neuritis phenotype. Nonetheless, the relations between epitopes on MOG protein remain unclear and need to be identified in the future. Full article
(This article belongs to the Special Issue Updates in Cell and Molecular Mechanisms of Autoimmune Diseases)
Show Figures

Figure 1

Figure 1
<p>Anti-MOG-IgG autoantibody-binding sites on the extracellular portion of MOG identified in patients with MOGAD.</p>
Full article ">
16 pages, 2690 KiB  
Article
Anti-Glycolipid Antibody Examination in Five EAE Models and Theiler’s Virus Model of Multiple Sclerosis: Detection of Anti-GM1, GM3, GM4, and Sulfatide Antibodies in Relapsing-Remitting EAE
by Kota Moriguchi, Yumina Nakamura, Ah-Mee Park, Fumitaka Sato, Motoi Kuwahara, Sundar Khadka, Seiichi Omura, Ijaz Ahmad, Susumu Kusunoki and Ikuo Tsunoda
Int. J. Mol. Sci. 2023, 24(16), 12937; https://doi.org/10.3390/ijms241612937 - 18 Aug 2023
Viewed by 2367
Abstract
Anti-glycolipid antibodies have been reported to play pathogenic roles in peripheral inflammatory neuropathies, such as Guillain–Barré syndrome. On the other hand, the role in multiple sclerosis (MS), inflammatory demyelinating disease in the central nervous system (CNS), is largely unknown, although the presence of [...] Read more.
Anti-glycolipid antibodies have been reported to play pathogenic roles in peripheral inflammatory neuropathies, such as Guillain–Barré syndrome. On the other hand, the role in multiple sclerosis (MS), inflammatory demyelinating disease in the central nervous system (CNS), is largely unknown, although the presence of anti-glycolipid antibodies was reported to differ among MS patients with relapsing-remitting (RR), primary progressive (PP), and secondary progressive (SP) disease courses. We investigated whether the induction of anti-glycolipid antibodies could differ among experimental MS models with distinct clinical courses, depending on induction methods. Using three mouse strains, SJL/J, C57BL/6, and A.SW mice, we induced five distinct experimental autoimmune encephalomyelitis (EAE) models with myelin oligodendrocyte glycoprotein (MOG)35–55, MOG92–106, or myelin proteolipid protein (PLP)139–151, with or without an additional adjuvant curdlan injection. We also induced a viral model of MS, using Theiler’s murine encephalomyelitis virus (TMEV). Each MS model had an RR, SP, PP, hyperacute, or chronic clinical course. Using the sera from the MS models, we quantified antibodies against 11 glycolipids: GM1, GM2, GM3, GM4, GD3, galactocerebroside, GD1a, GD1b, GT1b, GQ1b, and sulfatide. Among the MS models, we detected significant increases in four anti-glycolipid antibodies, GM1, GM3, GM4, and sulfatide, in PLP139–151-induced EAE with an RR disease course. We also tested cellular immune responses to the glycolipids and found CD1d-independent lymphoproliferative responses only to sulfatide with decreased interleukin (IL)-10 production. Although these results implied that anti-glycolipid antibodies might play a role in remissions or relapses in RR-EAE, their functional roles need to be determined by mechanistic experiments, such as injections of monoclonal anti-glycolipid antibodies. Full article
Show Figures

Figure 1

Figure 1
<p>Structures of representative glycolipids. The saccharide chains are composed of galactose, glucose, sialic acid, or N-acetyl-galactosamine, which bind ceramide. Ceramides are lipids composed of amino alcohol and fatty acid varying in length. Shown are 11 glycolipids that we used in the current study as antigens and determined anti-glycolipid antibody levels by enzyme-linked immunosorbent assays (ELISAs).</p>
Full article ">Figure 2
<p>Multiple sclerosis (MS) models with distinct clinical courses. (<b>A</b>) Myelin proteolipid protein (PLP)<sub>139–151</sub>-sensitized SJL/J mice developed relapsing-remitting (RR) experimental autoimmune encephalomyelitis (EAE). (<b>B</b>) PLP<sub>139–151</sub>-sensitized SJL/J mice with curdlan injection developed hyperacute EAE, and all mice died around 2 weeks post-induction (p.i.). (<b>C</b>) Myelin oligodendrocyte glycoprotein (MOG)<sub>92–106</sub>-sensitized SJL/J mice with curdlan injection developed primary progressive (PP) EAE without remission. (<b>D</b>) MOG<sub>92–106</sub>-sensitized A.SW mice developed chronic PP-EAE. (<b>E</b>) MOG<sub>35–55</sub>-sensitized C57BL/6 mice developed chronic EAE 1 month p.i. (<b>F</b>) Theiler’s murine encephalomyelitis virus (TMEV)-infected SJL/J mice developed a PP disease, TMEV-induced demyelinating disease (TMEV-IDD), around 1 month after virus inoculation (chronic phase). Although TMEV-infected mice also had neurological signs 1 week after infection (acute phase), the acute disease was induced by direct virus infection in the gray matter, leading to polioencephalomyelitis, not by demyelination in the white matter. Shown are representative disease courses.</p>
Full article ">Figure 3
<p>Anti-glycolipid antibody levels in MS models and untreated naïve mice (control). Using anti-mouse F(ab’)<sub>2</sub> antibody that detects all immunoglobulin (Ig) subclasses, we conducted ELISAs to examine the levels of antibodies against 10 glycolipids: GM1, GM2, GM3, GM4, GD3, galactocerebroside (GC), GD1a, GD1b, GT1b, and GQ1b. We measured the absorbances at 492 nm (Abs<sub>492nm</sub>) in glycolipid-coated and uncoated wells, and subtracted the absorbances of uncoated wells from those of glycolipid-coated wells. The data were the mean Abs<sub>492 nm</sub> + standard error of the mean (SEM) of serum samples. (<b>A</b>) Anti-glycolipid antibody levels of SJL/J mice. We detected higher anti-GM1, anti-GM3, and anti-GM4 antibodies in PLP<sub>139–151</sub>-sensitized EAE mice than the control mice (*, <span class="html-italic">p</span> &lt; 0.05; and **, <span class="html-italic">p</span> &lt; 0.01) (control, <span class="html-italic">n</span> = 8; PLP<sub>139–151</sub>, <span class="html-italic">n</span> = 8; MOG<sub>92–106</sub>, <span class="html-italic">n</span> = 6; and TMEV-IDD, <span class="html-italic">n</span> = 13). (<b>B</b>) Anti-glycolipid antibody levels of C57BL/6 mice. We did not detect significantly high anti-glycolipid antibodies compared with the control mice (control, <span class="html-italic">n</span> = 8; and MOG<sub>35–55</sub>, <span class="html-italic">n</span> = 8). (<b>C</b>) Anti-glycolipid antibody levels of A.SW mice. We did not detect significantly high anti-glycolipid antibodies compared with the control mice (control, <span class="html-italic">n</span> = 3; and MOG<sub>92–106</sub>, <span class="html-italic">n</span> = 8).</p>
Full article ">Figure 4
<p>Anti-sulfatide antibody and its subclass. (<b>A</b>) Anti-sulfatide antibodies were detected in all controls and MS models. SJL/J mice: control, <span class="html-italic">n</span> = 8; PLP<sub>139–151</sub>, <span class="html-italic">n</span> = 8; MOG<sub>92–106</sub>, <span class="html-italic">n</span> = 6; and TMEV-IDD, <span class="html-italic">n</span> = 13 (*, <span class="html-italic">p</span> &lt; 0.05; and **, <span class="html-italic">p</span> &lt; 0.01). C57BL/6 mice: control, <span class="html-italic">n</span> = 8; and MOG<sub>35–55</sub>, <span class="html-italic">n</span> = 8. A.SW mice: control, <span class="html-italic">n</span> = 3; and MOG<sub>92–106</sub>, <span class="html-italic">n</span> = 8. The data were the mean Abs<sub>492nm</sub> + SEM of serum samples. (<b>B</b>) Using sera from PLP<sub>139–151</sub>-sensitized SJL/J mice, we conducted ELISAs with anti-mouse IgM (●) or IgG (○) Fc-specific antibody as a detection antibody. Anti-sulfatide antibody was elevated at 14 days p.i. of EAE, and its subclass was IgM.</p>
Full article ">Figure 5
<p>Anti-glycolipid antibody levels and EAE severities. We collected serum samples on 14–21 days p.i. from PLP<sub>139–151</sub>-induced RR-EAE mice (<span class="html-italic">n</span> = 18) and evaluated the associations between anti-glycolipid antibody levels and EAE scores. Anti-GM1 (<b>A</b>), anti-GM3 (<b>B</b>), and anti-GM4 (<b>C</b>) antibody levels were undetectable or low in EAE mice with high disease severity (EAE score 3 or more). (<b>D</b>) Anti-sulfatide antibody levels were higher than the other anti-glycolipid antibodies, regardless of disease severity.</p>
Full article ">Figure 6
<p>Lymphoproliferative assays against glycolipids. We harvested mononuclear cells (MNCs) from PLP<sub>139–151</sub>-induced EAE mice. Using the Cell Counting Kit-8 (CCK-8), we examined lymphoproliferative responses in the presence or absence of glycolipids: GM1, GM3, GM4, GD1b, or sulfatide. To assess whether glycolipid antigens were presented on CD1d molecules, we conducted the assays in the presence (+) or absence (−) of anti-CD1d monoclonal antibody (mAb). (<b>A</b>) We observed higher lymphoproliferative responses to sulfatide, but not to the other glycolipids, compared with the vehicle control (**, <span class="html-italic">p</span> &lt; 0.01). Anti-CD1d mAb treatment did not affect the lymphoproliferative responses. The data were the mean + SEM of four pools of MNCs isolated from spleens and inguinal lymph nodes; one pool was from two to three mice. (<b>B</b>) We incubated MNCs with different doses of glycolipids and found significant lymphoproliferation only with sulfatide at 5 μg/mL, compared with the vehicle control (**, <span class="html-italic">p</span> &lt; 0.01). MNCs were isolated 3 weeks p.i. The data were the mean Abs<sub>450 nm</sub> ± SEM of two pools of MNCs. One pool was from spleens of two to three mice. We conducted the assays, using triplicate wells, and determined the statistical differences by analysis of variance (ANOVA).</p>
Full article ">Figure 7
<p>Cytokine production from sulfatide-stimulated MNCs in PLP<sub>139–151</sub>-induced EAE. We isolated MNCs from PLP<sub>139–151</sub>-induced EAE mice and incubated MNCs in the presence or absence of sulfatide. We quantified the amounts of interleukin (IL)-4, IL-10, IL-17, and interferon (IFN)-γ in the supernatants. We found that IL-10 production was suppressed by an addition of sulfatide, although it did not reach a statistical difference. IL-4 and IFN-γ were not detectable (N.D.). The data were the mean + SEM of four pools of spleen and inguinal lymph nodes from two to three mice.</p>
Full article ">
53 pages, 9253 KiB  
Article
Low Doses of β-Caryophyllene Reduced Clinical and Paraclinical Parameters of an Autoimmune Animal Model of Multiple Sclerosis: Investigating the Role of CB2 Receptors in Inflammation by Lymphocytes and Microglial
by Vahid Reza Askari, Vafa Baradaran Rahimi and Reza Shafiee-Nick
Brain Sci. 2023, 13(7), 1092; https://doi.org/10.3390/brainsci13071092 - 19 Jul 2023
Cited by 6 | Viewed by 2972
Abstract
Multiple Sclerosis (MS) is a prevalent inflammatory disease in which the immune system plays an essential role in the damage, inflammation, and demyelination of central nervous system neurons (CNS). The cannabinoid receptor type 2 (CB2) agonists possess anti-inflammatory effects against noxious [...] Read more.
Multiple Sclerosis (MS) is a prevalent inflammatory disease in which the immune system plays an essential role in the damage, inflammation, and demyelination of central nervous system neurons (CNS). The cannabinoid receptor type 2 (CB2) agonists possess anti-inflammatory effects against noxious stimuli and elevate the neuronal survival rate. We attempted to analyze the protective impact of low doses of β-Caryophyllene (BCP) in experimental autoimmune encephalomyelitis (EAE) mice as a chronic MS model. Immunization of female C57BL/6 mice was achieved through two subcutaneous injections into different areas of the hind flank with an emulsion that consisted of myelin Myelin oligodendrocyte glycoprotein (MOG)35–55 (150 µg) and complete Freund’s adjuvant (CFA) (400 µg) with an equal volume. Two intraperitoneal (i.p.) injections of pertussis toxin (300 ng) were performed on the animals on day zero (immunizations day) and 48 h (2nd day) after injection of MOG + CFA. The defensive effect of low doses of BCP (2.5 and 5 mg/kg/d) was investigated in the presence and absence of a CB2 receptor antagonist (1 mg/kg, AM630) in the EAE model. We also examined the pro/anti-inflammatory cytokine levels and the polarization of brain microglia and spleen lymphocytes in EAE animals. According to our findings, low doses of BCP offered protective impacts in the EAE mice treatment in a CB2 receptor-dependent way. In addition, according to results, BCP decreased the pathological and clinical defects in EAE mice via modulating adaptive (lymphocytes) and innate (microglia) immune systems from inflammatory phenotypes (M1/Th1/Th17) to anti-inflammatory (M2/Th2/Treg) phenotypes. Additionally, BCP elevated the anti-inflammatory cytokine IL-10 and reduced blood inflammatory cytokines. BCP almost targeted the systemic immune system more than the CNS immune system. Thus, a low dose of BCP can be suggested as a therapeutic effect on MS treatment with potent anti-inflammatory effects and possibly lower toxicity. Full article
(This article belongs to the Section Sleep and Circadian Neuroscience)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Treatment protocol, chemical structures, and flow cytometric analysis of cell-surface expression of receptors; (<b>A</b>) the summarized protocols of treatment and induction; (<b>B</b>) chemical configurations of the compounds used in the research.</p>
Full article ">Figure 2
<p><b>The effects of BCP treatment on body weight and clinical score of EAE mice;</b> (<b>A</b>) The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5 and 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) and BCP (5 mg/kg/day; p.o.) on the clinical score of EAE mice. (<b>B</b>) The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5 and 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) and BCP (5 mg/kg/day; p.o.) on the body weight of EAE mice. Data were expressed as mean ± SEM, n = 8 for experiment protocols. A two-way ANOVA test was performed with the following Tukey’s multiple comparisons tests. (<b><sup>+</sup></b>) makes a comparison between the vehicle and sham groups, <b><sup>+++</sup></b>: <span class="html-italic">p</span> &lt; 0.001.; (*) indicates the comparison with the vehicle group, ***: <span class="html-italic">p</span> &lt; 0.001; (<b><sup>#</sup></b>) makes a comparison between BCP and the DMF group in each graph, <b><sup>###</sup></b>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>The effects of treatment with BCP on the inflammatory cell infiltration into the spinal cords and pathological score with H&amp;E for enumerating infiltrating leukocytes; (<b>A</b>) The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) with BCP (5 mg/kg/day; p.o.) on the pathological score in EAE mice. (<b>B</b>) Sham group. (<b>C</b>) Vehicle. (<b>D</b>) DMF 60 mg/kg. (<b>E</b>) BCP 2.5 mg/kg. (<b>F</b>) BCP 5 mg/kg. (<b>G</b>) BCP 5 mg/kg + AM630; indicates lumbar spinal cord parts of H&amp;E staining (100×); 0—without inflammation; 1—low inflammatory cell levels, 2—perivascular infiltrates; and 3—the extended intensity of perivascular cuffing with extension into contiguous tissue. Data were presented as mean ± range, n = seven animals per group, three to four fields of view per animal for each experiment protocol. Considering that data has non-parametric nature, data analysis was performed using the Kruskal–Wallis test and Dunn’s post hoc multiple comparisons test. (*) indicates the comparison between varying doses of BCP and DMF groups and the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, and ***: <span class="html-italic">p</span> &lt; 0.001.; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001.; <sup>#</sup> makes comparisons between BCP to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3 Cont.
<p>The effects of treatment with BCP on the inflammatory cell infiltration into the spinal cords and pathological score with H&amp;E for enumerating infiltrating leukocytes; (<b>A</b>) The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) with BCP (5 mg/kg/day; p.o.) on the pathological score in EAE mice. (<b>B</b>) Sham group. (<b>C</b>) Vehicle. (<b>D</b>) DMF 60 mg/kg. (<b>E</b>) BCP 2.5 mg/kg. (<b>F</b>) BCP 5 mg/kg. (<b>G</b>) BCP 5 mg/kg + AM630; indicates lumbar spinal cord parts of H&amp;E staining (100×); 0—without inflammation; 1—low inflammatory cell levels, 2—perivascular infiltrates; and 3—the extended intensity of perivascular cuffing with extension into contiguous tissue. Data were presented as mean ± range, n = seven animals per group, three to four fields of view per animal for each experiment protocol. Considering that data has non-parametric nature, data analysis was performed using the Kruskal–Wallis test and Dunn’s post hoc multiple comparisons test. (*) indicates the comparison between varying doses of BCP and DMF groups and the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, and ***: <span class="html-italic">p</span> &lt; 0.001.; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001.; <sup>#</sup> makes comparisons between BCP to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of anti-inflammatory and pro-inflammatory cytokines in the EAE mice serums; (<b>A</b>) TNF-α, (<b>B</b>) IL-6; (<b>C</b>) IL-17; (<b>D</b>) IL-10; and (<b>E</b>) IL-17/IL-10 ratio. Data were presented as Mean ± SEM, n = 6 animals in each group per experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes comparisons between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of anti-inflammatory and pro-inflammatory cytokines in the EAE mice serums; (<b>A</b>) TNF-α, (<b>B</b>) IL-6; (<b>C</b>) IL-17; (<b>D</b>) IL-10; and (<b>E</b>) IL-17/IL-10 ratio. Data were presented as Mean ± SEM, n = 6 animals in each group per experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes comparisons between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of anti-inflammatory and pro-inflammatory cytokines in the EAE mice serums; (<b>A</b>) TNF-α, (<b>B</b>) IL-6; (<b>C</b>) IL-17; (<b>D</b>) IL-10; and (<b>E</b>) IL-17/IL-10 ratio. Data were presented as Mean ± SEM, n = 6 animals in each group per experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes comparisons between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on cell proliferation, levels of anti- and pro-inflammatory cytokines in spleen lymphocytes of EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) IL-6; (<b>D</b>) IFN-γ; (<b>E</b>) IL-17; (<b>F</b>) IL-4; (<b>G</b>) IL-10; (<b>H</b>) TGF-β<sub>1</sub>; (<b>I</b>) ratio of IFN-γ/IL-4; (<b>J</b>) IL-ratio of 17/IL-10; and (<b>K</b>) ratio of IL-17/ TGF-β<sub>1</sub>. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP and DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes the comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison with to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of transcription factors, including GATA3 (Th<sub>2</sub>), T-bet (Th<sub>1</sub>), Foxp3 (T<sub>reg</sub>), and ROR-γt (Th<sub>17</sub>) of lymphocytes isolated from EAE mice; (<b>A</b>) T-bet; (<b>B</b>) GATA3; (<b>C</b>) ROR-γt; (<b>D</b>) Foxp3; (<b>E</b>) ratio of T-bet/GATA3 (Th<sub>1</sub>/Th<sub>2</sub>); and (<b>F</b>) ratio of ROR-γt/Foxp3 (Th<sub>17</sub>/T<sub>reg</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01 and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> compared to DMF groups in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of transcription factors, including GATA3 (Th<sub>2</sub>), T-bet (Th<sub>1</sub>), Foxp3 (T<sub>reg</sub>), and ROR-γt (Th<sub>17</sub>) of lymphocytes isolated from EAE mice; (<b>A</b>) T-bet; (<b>B</b>) GATA3; (<b>C</b>) ROR-γt; (<b>D</b>) Foxp3; (<b>E</b>) ratio of T-bet/GATA3 (Th<sub>1</sub>/Th<sub>2</sub>); and (<b>F</b>) ratio of ROR-γt/Foxp3 (Th<sub>17</sub>/T<sub>reg</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01 and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> compared to DMF groups in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of transcription factors, including GATA3 (Th<sub>2</sub>), T-bet (Th<sub>1</sub>), Foxp3 (T<sub>reg</sub>), and ROR-γt (Th<sub>17</sub>) of lymphocytes isolated from EAE mice; (<b>A</b>) T-bet; (<b>B</b>) GATA3; (<b>C</b>) ROR-γt; (<b>D</b>) Foxp3; (<b>E</b>) ratio of T-bet/GATA3 (Th<sub>1</sub>/Th<sub>2</sub>); and (<b>F</b>) ratio of ROR-γt/Foxp3 (Th<sub>17</sub>/T<sub>reg</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01 and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> compared to DMF groups in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of transcription factors, including GATA3 (Th<sub>2</sub>), T-bet (Th<sub>1</sub>), Foxp3 (T<sub>reg</sub>), and ROR-γt (Th<sub>17</sub>) of lymphocytes isolated from EAE mice; (<b>A</b>) T-bet; (<b>B</b>) GATA3; (<b>C</b>) ROR-γt; (<b>D</b>) Foxp3; (<b>E</b>) ratio of T-bet/GATA3 (Th<sub>1</sub>/Th<sub>2</sub>); and (<b>F</b>) ratio of ROR-γt/Foxp3 (Th<sub>17</sub>/T<sub>reg</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01 and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> compared to DMF groups in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of transcription factors, including GATA3 (Th<sub>2</sub>), T-bet (Th<sub>1</sub>), Foxp3 (T<sub>reg</sub>), and ROR-γt (Th<sub>17</sub>) of lymphocytes isolated from EAE mice; (<b>A</b>) T-bet; (<b>B</b>) GATA3; (<b>C</b>) ROR-γt; (<b>D</b>) Foxp3; (<b>E</b>) ratio of T-bet/GATA3 (Th<sub>1</sub>/Th<sub>2</sub>); and (<b>F</b>) ratio of ROR-γt/Foxp3 (Th<sub>17</sub>/T<sub>reg</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01 and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> compared to DMF groups in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of transcription factors, including GATA3 (Th<sub>2</sub>), T-bet (Th<sub>1</sub>), Foxp3 (T<sub>reg</sub>), and ROR-γt (Th<sub>17</sub>) of lymphocytes isolated from EAE mice; (<b>A</b>) T-bet; (<b>B</b>) GATA3; (<b>C</b>) ROR-γt; (<b>D</b>) Foxp3; (<b>E</b>) ratio of T-bet/GATA3 (Th<sub>1</sub>/Th<sub>2</sub>); and (<b>F</b>) ratio of ROR-γt/Foxp3 (Th<sub>17</sub>/T<sub>reg</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, **: <span class="html-italic">p</span> &lt; 0.01 and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> compared to DMF groups in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of cell proliferation, anti-inflammatory and inflammatory cytokines of microglia separated from EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) PGE<sub>2</sub>; and (<b>D</b>) IL-10. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates a comparison between varying doses of BCP or DMF groups to the vehicle group, ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of cell proliferation, anti-inflammatory and inflammatory cytokines of microglia separated from EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) PGE<sub>2</sub>; and (<b>D</b>) IL-10. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates a comparison between varying doses of BCP or DMF groups to the vehicle group, ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of cell proliferation, anti-inflammatory and inflammatory cytokines of microglia separated from EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) PGE<sub>2</sub>; and (<b>D</b>) IL-10. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates a comparison between varying doses of BCP or DMF groups to the vehicle group, ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on the levels of cell proliferation, anti-inflammatory and inflammatory cytokines of microglia separated from EAE mice; (<b>A</b>) Cell proliferation; (<b>B</b>) TNF-α; (<b>C</b>) PGE<sub>2</sub>; and (<b>D</b>) IL-10. Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates a comparison between varying doses of BCP or DMF groups to the vehicle group, ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05 and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on intracellular levels of Arg-1 and iNOS and their products, urea, and NO of microglia isolated from EAE mice; (<b>A</b>) iNOS; (<b>B</b>) Arg-1; (<b>C</b>) ratio of iNOS/Arg-1 (M<sub>1</sub>/M<sub>2</sub>); (<b>D</b>) NO; (<b>E</b>) Urea; (<b>F</b>) ratio of NO/Urea (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on intracellular levels of Arg-1 and iNOS and their products, urea, and NO of microglia isolated from EAE mice; (<b>A</b>) iNOS; (<b>B</b>) Arg-1; (<b>C</b>) ratio of iNOS/Arg-1 (M<sub>1</sub>/M<sub>2</sub>); (<b>D</b>) NO; (<b>E</b>) Urea; (<b>F</b>) ratio of NO/Urea (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on intracellular levels of Arg-1 and iNOS and their products, urea, and NO of microglia isolated from EAE mice; (<b>A</b>) iNOS; (<b>B</b>) Arg-1; (<b>C</b>) ratio of iNOS/Arg-1 (M<sub>1</sub>/M<sub>2</sub>); (<b>D</b>) NO; (<b>E</b>) Urea; (<b>F</b>) ratio of NO/Urea (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on intracellular levels of Arg-1 and iNOS and their products, urea, and NO of microglia isolated from EAE mice; (<b>A</b>) iNOS; (<b>B</b>) Arg-1; (<b>C</b>) ratio of iNOS/Arg-1 (M<sub>1</sub>/M<sub>2</sub>); (<b>D</b>) NO; (<b>E</b>) Urea; (<b>F</b>) ratio of NO/Urea (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on intracellular levels of Arg-1 and iNOS and their products, urea, and NO of microglia isolated from EAE mice; (<b>A</b>) iNOS; (<b>B</b>) Arg-1; (<b>C</b>) ratio of iNOS/Arg-1 (M<sub>1</sub>/M<sub>2</sub>); (<b>D</b>) NO; (<b>E</b>) Urea; (<b>F</b>) ratio of NO/Urea (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8 Cont.
<p>The effects of DMF (60 mg/kg; 30 mg/kg twice a day; p.o.), BCP (2.5, 5 mg/kg/day; p.o.), and the combination of a CB<sub>2</sub> antagonist AM630 (1 mg/kg; i.p.) with BCP (5 mg/kg/day; p.o.) on intracellular levels of Arg-1 and iNOS and their products, urea, and NO of microglia isolated from EAE mice; (<b>A</b>) iNOS; (<b>B</b>) Arg-1; (<b>C</b>) ratio of iNOS/Arg-1 (M<sub>1</sub>/M<sub>2</sub>); (<b>D</b>) NO; (<b>E</b>) Urea; (<b>F</b>) ratio of NO/Urea (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM, n = six animals per group for each experiment protocol. Two-way ANOVA and Tukey’s multiple comparisons test were performed. (*) indicates the comparison between varying doses of BCP or DMF groups to the vehicle group, *: <span class="html-italic">p</span> &lt; 0.05, **: <span class="html-italic">p</span> &lt; 0.01, and ***: <span class="html-italic">p</span> &lt; 0.001; (<sup>+</sup>) makes a comparison between the vehicle and sham groups, <sup>+++</sup>: <span class="html-italic">p</span> &lt; 0.001; <sup>#</sup> in comparison to DMF group in each graph, <sup>#</sup>: <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup>: <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup>: <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 9
<p>The effects of treatment with BCP on the levels of CNS immunity (microglia) and systemic immunity (blood and spleen lymphocytes) in EAE mice; each column presents the effects of DMF, BCP, and the combination of AM630 and BCP on the decreased levels of anti-inflammatory/ inflammatory ratios in the spleen lymphocytes (Th<sub>17</sub>/T<sub>reg</sub> and Th<sub>1</sub>/Th<sub>2</sub>), blood (IL-17/IL-10), and microglia (M<sub>1</sub>/M<sub>2</sub>). Data were presented as Mean ± SEM. The B, T<sub>17</sub>, T<sub>1</sub>, and M<sub>1</sub> Abbreviations represent the ratios of blood IL-17/IL-10, Th<sub>17</sub>/T<sub>reg</sub>, Th<sub>1</sub>/Th<sub>2</sub> and M<sub>1</sub>/M<sub>2</sub>.</p>
Full article ">
7 pages, 1236 KiB  
Case Report
Isolated Myelopathy in Occult Breast Carcinoma with Negative Paraneoplastic Antibodies: A Case Report of a Rare Condition
by Loredana Paciolla, Giulia Galli, Domizia Vecchio, Samuel Padelli, Cristoforo Comi, Roberto Cantello and Eleonora Virgilio
Sclerosis 2023, 1(1), 60-66; https://doi.org/10.3390/sclerosis1010007 - 18 May 2023
Viewed by 1886
Abstract
Isolated paraneoplastic myelopathy (IPM) is a rare neurological manifestation of systemic cancer and represents an intermediate-risk phenotype of disease according to the diagnostic criteria for Paraneoplastic Neurologic Syndromes (PNS). Here, we present the case of a 47-year-old woman who developed subacute cervical myelopathy [...] Read more.
Isolated paraneoplastic myelopathy (IPM) is a rare neurological manifestation of systemic cancer and represents an intermediate-risk phenotype of disease according to the diagnostic criteria for Paraneoplastic Neurologic Syndromes (PNS). Here, we present the case of a 47-year-old woman who developed subacute cervical myelopathy and was then diagnosed with breast cancer. Through this lens, we provide a discussion of current literature on IPM. Over four months, our patient developed progressive tetraparesis, hypoesthesia with C3 level, and urinary retention. The first MRI was negative, but a four-month-control MRI showed a T2-hyperintense spinal lesion (C2–C7 and T2–T4). Cerebrospinal fluid (CSF) analysis was normal. Infective and autoimmune screening, including onconeural, anti-MOG, and aquaporin-4 antibodies, was unremarkable. The total-body CT scan was negative, but total-body PET-CT scan evidenced an enlarged axillary lymph node, with the detection of breast cancer cells at fine-needle aspiration. Despite negative mammography, a breast MRI confirmed a mammary nodule, which was removed, and a ductal infiltrating breast carcinoma diagnosis was made. Her neurological condition partially improved after steroid therapy. Our final diagnosis was probable IPM, according to PNS criteria. This rare condition affects most frequently middle-aged women and is often associated with breast and lung cancer, even if two-thirds of patients’ cancer diagnosis is subsequent to the onset of neurological deficits. Clinical presentation is often subtle, and CSF analysis, neuroimaging, and onconeural autoantibodies could be negative or non-specific. However, if the suspect of paraneoplastic disease is strong, cancer should be searched thoroughly since early diagnosis and treatment are associated with a better outcome. Full article
Show Figures

Figure 1

Figure 1
<p>Sagittal spinal cord MRI T2-weighted image showing a longitudinally extensive T2 signal abnormality from C2 to C7 (star) with spinal cord swelling particularly at C3 levels in the dorsal column. After administration of gadolinium no pathological contrast enhancement is appreciated. Radiological findings are consistent with inflammatory myelitis.</p>
Full article ">Figure 2
<p>Whole-body PET scan revealing an enlarged axillary lymph node (red star), with fluorodeoxyglucose hyperaccumulation (in (<bold>a</bold>) an axial image (<bold>b</bold>) a sagittal image and (<bold>c</bold>) a frontal section). No spinal cord or breast hypermetabolic foci were found. The letter correspond to axial (A), sagittal (B) and frontal (H).</p>
Full article ">Figure 3
<p>The most recent diagnostic criteria for PNS defines three levels of certainty of diagnosis (possible, probable, and definite PNS) based on clinical phenotype (high risk or intermediate risk phenotype or phenotype not associated with cancer), the presence or absence of antibodies, and the type of antibodies and the presence or absence of cancer. We obtain a score ranging from 0–10 (&lt;3 not PNS, 4–5 possible PNS, 6–7 probable PNS, &gt;8 definite PNS). Our case scored 6, corresponding to a probable PNS. Figure created with Biorender.com (Accessed on 21 November 2022).</p>
Full article ">
31 pages, 7526 KiB  
Article
EAE of Mice: Enzymatic Cross Site-Specific Hydrolysis of H2A Histone by IgGs against H2A, H1, H2B, H3, and H4 Histones and Myelin Basic Protein
by Andrey E. Urusov, Kseniya S. Aulova, Pavel S. Dmitrenok, Valentina N. Buneva and Georgy A. Nevinsky
Int. J. Mol. Sci. 2023, 24(10), 8636; https://doi.org/10.3390/ijms24108636 - 12 May 2023
Viewed by 1642
Abstract
Histones play vital roles in chromatin function and gene transcription; however, they are very harmful in the intercellular space because they stimulate systemic inflammatory and toxic responses. Myelin basic protein (MBP) is the major protein of the axon myelin–proteolipid sheath. Antibodies–abzymes with various [...] Read more.
Histones play vital roles in chromatin function and gene transcription; however, they are very harmful in the intercellular space because they stimulate systemic inflammatory and toxic responses. Myelin basic protein (MBP) is the major protein of the axon myelin–proteolipid sheath. Antibodies–abzymes with various catalytic activities are specific features of some autoimmune diseases. IgGs against individual histones (H2A, H1, H2B, H3, and H4) and MBP were isolated from the blood of experimental-autoimmune-encephalomyelitis-prone C57BL/6 mice by several affinity chromatographies. These Abs–abzymes corresponded to various stages of EAE development: spontaneous EAE, MOG, and DNA–histones accelerated the onset, acute, and remission stages. IgGs-abzymes against MBP and five individual histones showed unusual polyreactivity in the complex formation and enzymatic cross-reactivity in the specific hydrolysis of the H2A histone. All the IgGs of 3-month-old mice (zero time) against MBP and individual histones demonstrated from 4 to 35 different H2A hydrolysis sites. The spontaneous development of EAE over 60 days led to a significant change in the type and number of H2A histone hydrolysis sites by IgGs against five histones and MBP. Mice treatment with MOG and the DNA–histone complex changed the type and number of H2A hydrolysis sites compared to zero time. The minimum number (4) of different H2A hydrolysis sites was found for IgGs against H2A (zero time), while the maximum (35) for anti-H2B IgGs (60 days after mice treatment with DNA–histone complex). Overall, it was first demonstrated that at different stages of EAE evolution, IgGs–abzymes against individual histones and MBP could significantly differ in the number and type of specific sites of H2A hydrolysis. The possible reasons for the catalytic cross-reactivity and great differences in the number and type of histone H2A cleavage sites were analyzed. Full article
(This article belongs to the Collection Feature Paper Collection in Biochemistry)
Show Figures

Figure 1

Figure 1
<p>SDS-PAGE analysis of H2A histone hydrolysis by IgGs–abzymes against five histones (<b>A</b>) and this histone with IgGs against MBP (<b>B</b>) as well as splitting myelin basic protein by IgGs against five histones (<b>C</b>) and IgGs–abzymes against MBP (<b>D</b>). Lane C corresponds to the histones (<b>A</b>,<b>B</b>) and MBP (<b>C</b>,<b>D</b>) incubated without IgGs. MBP and a mixture of five histones with and without IgGs (0.03 mg/mL) were incubated for 14 h.</p>
Full article ">Figure 2
<p>MALDI spectra corresponding to products of H2A histone (0.85 mg/mL) hydrolysis in the absence (<b>A</b>) and the presence of IgGs (0.04 mg/mL) against three histones: Con-aH2A-0d (<b>B</b>), Spont-aH2A-60d (<b>C</b>), Con-aH1-0d (<b>D</b>), Spont-aH1-60d (<b>E</b>), Con-aH2B-0d (<b>F</b>), and Spont-aHB-60d (<b>G</b>). All designations of IgG preparations and the values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the Figure.</p>
Full article ">Figure 3
<p>MALDI spectra corresponding to products of H2A histone (0.85 mg/mL) hydrolysis in the presence of IgGs (0.04 mg/mL) against two (H3 and H4) histones: Con-aH3-0d (<b>A</b>), Spont-aH3-60d (<b>B</b>), Con-aH4-0d (<b>C</b>), and Spont-aH4-60d (<b>D</b>). All designations of IgG preparations and the values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the Figure.</p>
Full article ">Figure 4
<p>MALDI spectra of H2A (0.85 mg/mL) hydrolysis products with IgGs (0.04 mg/mL) against five histones corresponding to 20 days after mice immunization with MOG: MOG20-aH2A (<b>A</b>), MOG20-aH1 (<b>B</b>), MOG20-aH2B (<b>C</b>), MOG20-aH3 (<b>D</b>), and MOG20-aH4 (<b>E</b>). All designations of IgG preparations and the values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the Figure.</p>
Full article ">Figure 5
<p>MALDI spectra of H2A (0.85 mg/mL) hydrolysis products with IgGs (0.04 mg/mL) against three histones corresponding to 20 and 60 days after mice immunization with DNA–histone complex: DNA20-aH2A (<b>A</b>), DNA60-aH2A (<b>B</b>), DNA20-aH1 (<b>C</b>), DNA60-aH1 (<b>D</b>), DNA20-aH2B (<b>E</b>), and DNA60-aH2B (<b>F</b>). All designations of IgG preparations and the values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the Figure.</p>
Full article ">Figure 6
<p>MALDI spectra of H2A (0.85 mg/mL) hydrolysis products with IgGs (0.04 mg/mL) against two H3 and H4 histones and MBP corresponding to 20 and 60 days after mice immunization with DNA–histone complex: DNA20-aH3 (<b>A</b>), DNA60-aH3 (<b>B</b>), DNA20-aH4 (<b>C</b>), DNA60-aH4 (<b>D</b>), DNA20-aMBP (<b>E</b>), and DNA20-aMBP (<b>F</b>) and DNA60-aMBP (<b>G</b>). All designations of IgG preparations and the values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the Figure.</p>
Full article ">Figure 7
<p>Sites of H2A hydrolysis (in red) by IgGs against three histones (H2A, H1, and H2B) corresponding to zero time (3-month-old mice) and after spontaneous development of EAE (before mice immunization) over 60 days: Con-aH1-0d (<b>A</b>), Spont-aH1-60d (<b>B</b>), Con-aH2A-0d (<b>C</b>), Spont-aH2A-60d (<b>D</b>), Con-aH2B-0d (<b>E</b>), and Spont-aH2B-60d (<b>F</b>). Major sites of H2A cleavage are shown by stars (★), moderate ones by arrows (↓), and minor sites of the cleavages by small stars (*).</p>
Full article ">Figure 8
<p>Sites of H2A hydrolysis (in red) by IgGs against H3 and H4 histones corresponding to zero time (3-month-old mice) and after spontaneous development of EAE (before mice immunization) over 60 days: Con-aH3-0d (<b>A</b>), Spont-aH3-60d (<b>B</b>), Con-aH4-0d (<b>C</b>), and Spont-aH4-60d (<b>D</b>). Major sites of H2A cleavage are shown by stars (★), moderate ones by arrows (↓), and minor sites of the cleavages by small stars (*).</p>
Full article ">Figure 9
<p>Sites of H2A hydrolysis (in red) by IgGs against five H1–H4 histones corresponding to 20 days after mice immunization with MOG: MOG20-aH2A (<b>A</b>), MOG20-aH1 (<b>B</b>), MOG20-aH2B (<b>C</b>), MOG20-aH3 (<b>D</b>), and MOG20-aH4 (<b>E</b>). Major sites of H2A cleavage are shown by stars (★), moderate ones by arrows (↓), and minor sites of the cleavages by small stars (*).</p>
Full article ">Figure 10
<p>Sites of H2A hydrolysis (in red) by IgGs against five individual H1–H4 histones corresponding to 20 days after mice immunization with DNA–histone complex: DNA20-aH2A (<b>A</b>), DNA20-aH1 (<b>B</b>), DNA20-aH2B (<b>C</b>), DNA20-aH3 (<b>D</b>), and DNA20-aH4 (<b>E</b>). Major sites of H2A cleavage are shown by stars (★), moderate ones by arrows (↓), and minor sites of the cleavages by small stars (*).</p>
Full article ">Figure 11
<p>Sites of H2A hydrolysis (in red) by IgGs against five H1–H4 histones corresponding to 60 days after mice immunization with DNA–histone complex: DNA60-aH2A (<b>A</b>), DNA60-aH1 (<b>B</b>), DNA60-aH2B (<b>C</b>), DNA60-aH3 (<b>D</b>), and DNA60-aH4 (<b>E</b>). Major sites of H2A cleavage are shown by stars (★), moderate ones by arrows (↓), and minor sites of the cleavages by small stars (*).</p>
Full article ">Figure 12
<p>Sites of H2A hydrolysis (in red) by IgGs against MBP corresponding to zero time (3-month-old mice), 20 days after mice treatment with MOG as well as 20 and 60 days after mice immunization with DNA–histone complex: Con-aMBP-0d (<b>A</b>), MOG20-aMBP (<b>B</b>), DNA20-aMBP (<b>C</b>), and DNA60-aMBP (<b>D</b>). Major sites of H2A cleavage are shown by stars (★), moderate ones by arrows (↓), and minor sites of the cleavages by small stars (*).</p>
Full article ">
30 pages, 4510 KiB  
Article
EAE of Mice: Enzymatic Cross Site-Specific Hydrolysis of H2B Histone by IgGs against H1, H2A, H2B, H3, and H4 Histones and Myelin Basic Protein
by Andrey E. Urusov, Kseniya S. Aulova, Pavel S. Dmitrenok, Valentina N. Buneva and Georgy A. Nevinsky
Molecules 2023, 28(7), 2973; https://doi.org/10.3390/molecules28072973 - 27 Mar 2023
Cited by 2 | Viewed by 1744
Abstract
Histones have vital roles in chromatin functioning and gene transcription. At the same time, they are pernicious in intercellular space because they stimulate systemic inflammatory and toxic responses. Myelin basic protein (MBP) is the major protein of the axon myelin–proteolipid sheath. Antibody-abzymes with [...] Read more.
Histones have vital roles in chromatin functioning and gene transcription. At the same time, they are pernicious in intercellular space because they stimulate systemic inflammatory and toxic responses. Myelin basic protein (MBP) is the major protein of the axon myelin–proteolipid sheath. Antibody-abzymes with various catalytic activities are specific features of some autoimmune diseases. IgGs against five individual histones (H2B, H1, H2A, H3, and H4) and MBP were isolated from the blood of experimental autoimmune encephalomyelitis-prone C57BL/6 mice by affinity chromatography. Abzymes corresponding to various stages of EAE development, including spontaneous EAE, myelin oligodendrocyte glycoprotein (MOG)- and DNA-histone complex-accelerated onset, as well as acute and remission stages, were analyzed. IgG-abzymes against MBP and five individual histones showed unusual polyreactivity in complex formation and enzymatic cross-reactivity in the specific hydrolysis of H2B histone. All IgGs against MBP and individual histones in 3-month-old mice (zero time) demonstrated from 4 to 11 different H2B hydrolysis sites. Spontaneous development of EAE during 60 days led to a significant change in the type and number of H2B hydrolysis sites by IgGs against the five histones and MBP. Mouse treatment with MOG and DNA-histone complex changed the type and number of H2B hydrolysis sites compared to zero time. The minimum number (3) of different H2B hydrolysis sites was found for IgGs against H3 20 days after mouse immunization with DNA-histone complex, whereas the maximum number (33) for anti-H2B IgGs was found 60 days after mouse treatment with DNA-histone complex. Overall, this is the first study to demonstrate that at different stages of EAE evolution, IgG-abzymes against five individual histones and MBP could significantly differ in the specific sites and number of H2B hydrolysis sites. Possible reasons for the catalytic cross-reactivity and significant differences in the number and type of histone H2B cleavage sites were analyzed. Full article
Show Figures

Figure 1

Figure 1
<p>SDS-PAGE analysis of H2B (13.8 kDa) hydrolysis by IgGs against the five histones (<b>A</b>) and Abs against MBP (<b>B</b>) as well as splitting myelin basic protein (18.5 kDa) by antibodies against the five histones (<b>C</b>) and IgG-abzymes against MBP (<b>D</b>). Lanes C correspond to the histones (<b>A</b>,<b>B</b>) and MBP (<b>C</b>,<b>D</b>) incubated without IgG-abzymes. MBP and a mixture of five histones with and without IgGs (0.03 mg/mL) were incubated for 14 h. The full gel pictures are given in <a href="#app1-molecules-28-02973" class="html-app">Supplementary Data</a> (addition to <a href="#molecules-28-02973-f001" class="html-fig">Figure 1</a>).</p>
Full article ">Figure 2
<p>MALDI spectra corresponding to products of H2B histone (0.9 mg/mL) hydrolysis in the absence (<b>A</b>) or presence of IgGs (0.04 mg/mL) against three histones: control (H2B incubated without Abs) (<b>A</b>), Con-aH1-0d (<b>B</b>), Spont-aH1-60d (<b>C</b>), Con-aH2A-0d (<b>D</b>), Spont-aH2A-60d (<b>E</b>), Con-aH2B-0d (<b>F</b>), and Spont-aHB-60d (<b>G</b>). All designations of IgG preparations and values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the figure.</p>
Full article ">Figure 3
<p>MALDI spectra corresponding to products of H2B histone (0.9 mg/mL) hydrolysis in the presence of IgGs (0.04 mg/mL) against two histones corresponding to zero time and after spontaneous development of EAE during 60 days: Con-aH3-0d (<b>A</b>), Spont-aH3-60d (<b>B</b>), Con-aH4-0d (<b>C</b>), and Spont-aH4-60d (<b>D</b>). All designations of IgG preparations and values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the figure.</p>
Full article ">Figure 4
<p>MALDI spectra of H2B (0.9 mg/mL) hydrolysis products with IgGs against the five histones corresponding to 20 days after mouse immunization with MOG: MOG20-aH1 (<b>A</b>), MOG20-aH2A (<b>B</b>), MOG20-aH2B (<b>C</b>), MOG20-aH3 (<b>D</b>), and MOG20-aH4 (<b>E</b>). All designations of IgG preparations and values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the figure.</p>
Full article ">Figure 5
<p>MALDI spectra of H2B (0.9 mg/mL) hydrolysis products with IgGs against three histones corresponding to 20 and 60 days after mouse immunization with DNA-histone complex: DNA20-aH1 (<b>A</b>), DNA60-aH1 (<b>B</b>), DNA20-aH2A (<b>C</b>), DNA60-aH2A (<b>D</b>), DNA20-aH2B (<b>E</b>), and DNA60-aH2B (<b>F</b>). All designations of IgG preparations and values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the figure.</p>
Full article ">Figure 6
<p>MALDI spectra of H2B (0.9 mg/mL) hydrolysis products with IgGs against two histones and MBP corresponding to 20 and 60 days after mouse immunization with DNA-histone complex: DNA20-aH3 (<b>A</b>), DNA60-aH3 (<b>B</b>), DNA20-aH4 (<b>C</b>), DNA60-aH4 (D), MOG20-aMBP (<b>E</b>), DNA20-aMBP (<b>F</b>), and DNA60-aMBP (<b>G</b>). All designations of IgG preparations and the values of <span class="html-italic">m</span>/<span class="html-italic">z</span> are shown in the Figure.</p>
Full article ">Figure 7
<p>Sites of H2B hydrolysis by IgGs against three histones (H1, H2A, and H2B) corresponding to zero time (3-month-old mice) and after spontaneous development of EAE (before mouse immunization) during 60 days: Con-aH1-0d (<b>A</b>), Spont-aH1-60d (<b>B</b>), Con-aH2A-0d (<b>C</b>), Spont-aH2A-60d (<b>D</b>), Con-aH2B-0d (<b>E</b>), and Spont-aH2B-60d (<b>F</b>). Major sites of H2B cleavage are shown by stars (★), moderate sites by arrows (↓), and minor sites of cleavage by small stars (*).</p>
Full article ">Figure 8
<p>Sites of H2B hydrolysis by IgGs against H3 and H4 histones corresponding to zero time (3-month-old mice) and after spontaneous development of EAE (before mouse immunization) during 60 days: Con-aH3-0d (<b>A</b>), Spont-aH3-60d (<b>B</b>), Con-aH4-0d (<b>C</b>), and Spont-aH4-60d (<b>D</b>). Major sites of H2B cleavage are shown by stars (★), moderate sites by arrows (↓), and minor sites of cleavage by small stars (*).</p>
Full article ">Figure 9
<p>Sites of H2B hydrolysis by IgGs against five H1-H4 histones corresponding to 20 days after mouse immunization with MOG: MOG20-aH1 (<b>A</b>), MOG20-aH2A (<b>B</b>), MOG20-aH2B (<b>C</b>), MOG20-aH3 (<b>D</b>), and MOG20-aH4 (<b>E</b>). Major sites of H1 cleavage are shown by stars (★), moderate sites by arrows (↓), and minor sites of cleavage by small stars (*).</p>
Full article ">Figure 10
<p>Sites of H2B hydrolysis by IgGs against three individual H1-H2B histones corresponding to 20 and 60 days after mouse immunization with DNA-histone complex: DNA20-aH1 (<b>A</b>), DNA60-aH1 (<b>B</b>), DNA20-aH2A (<b>C</b>), DNA60-aH2A (<b>D</b>), DNA20-aH2B (<b>E</b>), and DNA60-aH2B (<b>F</b>). Major sites of H2A cleavage are shown by stars (★), moderate sites by arrows (↓), and minor sites of cleavage by small stars (*).</p>
Full article ">Figure 11
<p>Sites of H2B hydrolysis by IgGs against H3 and H4 histones corresponding to 20 and 60 days after mouse immunization with DNA-histone complex: DNA20-aH3 (<b>A</b>), DNA60-aH3 (<b>B</b>), DNA20-aH4 (<b>C</b>), DNA60-aH4 (<b>D</b>), and anti-MBP IgGs 20 days after mouse immunization with MOG-MOG20-aMBP (<b>E</b>), as well as 20 and 60 days after mouse immunization with DNA-histone complex DNA20-aMBP (<b>F</b>) and DNA60-aMBP (<b>G</b>). Major sites of H2A cleavage are shown by stars (★), moderate sites by arrows (↓), and minor sites of cleavage by small stars (*).</p>
Full article ">
8 pages, 3441 KiB  
Case Report
Anti-MOG Antibody-Associated Unilateral Cortical Encephalitis with Bilateral Meningeal Involvement: A Case Report
by Bo Ren, Shiying Li, Bin Liu, Jinxia Zhang and Yaqing Feng
Brain Sci. 2023, 13(2), 283; https://doi.org/10.3390/brainsci13020283 - 8 Feb 2023
Cited by 2 | Viewed by 2214
Abstract
A 27-year-old Han Chinese woman presented with fever, headache, lethargy, and difficulty in expression. Magnetic resonance imaging (MRI) detected extensive hyperintensity of the left-sided frontoparietal, temporal, occipital, and insular cortices via fluid-attenuated inversion recovery (FLAIR) imaging. Post-contrast MRI revealed linear enhancement in the [...] Read more.
A 27-year-old Han Chinese woman presented with fever, headache, lethargy, and difficulty in expression. Magnetic resonance imaging (MRI) detected extensive hyperintensity of the left-sided frontoparietal, temporal, occipital, and insular cortices via fluid-attenuated inversion recovery (FLAIR) imaging. Post-contrast MRI revealed linear enhancement in the frontoparietal, temporal, and occipital sulci bilaterally. The detection of anti-myelin oligodendrocyte glycoprotein (MOG) was positive in the cerebrospinal fluid (CSF) and serum. The patient was diagnosed with anti-MOG antibody-associated unilateral cortical encephalitis with bilateral meningeal involvement. The patient received low doses of intravenous dexamethasone followed by oral prednisone, which was tapered until withdrawal. The treatment significantly improved the patient’s symptoms. A one-month follow-up showed that the patient gradually resumed her normal lifestyle. No further relapse was recorded after a one-year follow-up. MRI performed almost a year after the initial symptom onset showed that the FLAIR signal had decreased in the left insular lobe, and the abnormal cortical signal of the FLAIR in the original left frontotemporal occipital lobe had disappeared. Thus, we report a rare case of anti-MOG antibody encephalitis (unilateral cortical encephalitis with bilateral meningeal involvement) in an adult patient. This study provides a reference for the clinical diagnosis and treatment of MOG antibody-associated unilateral cortical encephalitis. Full article
(This article belongs to the Section Molecular and Cellular Neuroscience)
Show Figures

Figure 1

Figure 1
<p>EEG showing the main rhythmic activities (low-amplitude α wave, irregular waveform, scattered sharp wave, and sharp–slow wave). The visual response did not significantly change (100 µ/cm and 30 mm/s).</p>
Full article ">Figure 2
<p>Brain MRI of the patient at admission (2 weeks after symptom onset) showing extensive hyperintensity of the left-sided frontoparietal, temporal, occipital, and insular cortices on FLAIR imaging (arrows) (<b>A</b>–<b>D</b>). Post-contrast MRI revealed linear enhancement in frontoparietal, temporal, and occipital sulci bilaterally (ellipses) (<b>E</b>–<b>H</b>). Post-contrast MRI after 2 weeks of treatment showed that the linear enhancement in these areas was less than before (ellipses) (<b>I</b>–<b>L</b>). MRI repeated after two months. The FLAIR signal had decreased in the left insular lobe, and the abnormal signal was partially absorbed in the left frontoparietal, temporal, and occipital cortices (arrows) (<b>M</b>–<b>P</b>). MRI performed almost one year after the initial symptom onset showed the FLAIR signal had decreased in the left insular lobe, and the abnormal cortical signal of FLAIR has disappeared in the original left frontal, temporal, parietal, and occipital lobes (<b>Q</b>–<b>T</b>).</p>
Full article ">
18 pages, 4594 KiB  
Article
Bacillus amyloliquifaciens-Supplemented Camel Milk Suppresses Neuroinflammation of Autoimmune Encephalomyelitis in a Mouse Model by Regulating Inflammatory Markers
by Hairul Islam Mohamed Ibrahim, Abdullah Sheikh, Hany Ezzat Khalil and Ashraf Khalifa
Nutrients 2023, 15(3), 550; https://doi.org/10.3390/nu15030550 - 20 Jan 2023
Cited by 8 | Viewed by 2718
Abstract
Multiple sclerosis (MS), a distinct autoimmune neuroinflammatory disorder, affects millions of people worldwide, including Saudi Arabia. Changes in the gut microbiome are linked to the development of neuroinflammation via mechanisms that are not fully understood. Prebiotics and probiotics in camel milk that has [...] Read more.
Multiple sclerosis (MS), a distinct autoimmune neuroinflammatory disorder, affects millions of people worldwide, including Saudi Arabia. Changes in the gut microbiome are linked to the development of neuroinflammation via mechanisms that are not fully understood. Prebiotics and probiotics in camel milk that has been fermented have a variety of health benefits. In this study, Bacillus amyloliquefaciens-supplemented camel milk (BASY) was used to assess its preventive effect on MS symptoms in a myelin oligodendrocyte glycoprotein (MOG)-immunized C57BL6J mice model. To this end, MOG-induced experimental autoimmune encephalomyelitis (EAE) was established and the level of disease index, pathological scores, and anti-inflammatory markers of BASY-treated mice using macroscopic and microscopic examinations, qPCR and immunoblot were investigated. The results demonstrate that BASY significantly reduced the EAE disease index, increased total microbial load (2.5 fold), and improved the levels of the short-chain fatty acids propionic, butyric and caproic acids in the diseased mice group. Additionally, myeloperoxidase (MPO) proinflammatory cytokines (IL-1β, IL-6, IL-17, TNF-α) and anti-inflammatory cytokines (TGF-β) were regulated by BASY treatment. Significant suppression of MPO and VCAM levels were noticed in the BASY-treated group (from 168 to 111 µM and from 34 to 27 pg/mL, respectively), in comparison to the EAE group. BASY treatment significantly reduced the expression of inflammatory cytokines, inflammatory progression related transcripts, and inflammatory progression protein markers. In conclusion, BASY significantly reduced the symptoms of EAE mice and may be used to develop a probiotic-based diet to promote host gut health. The cumulative findings of this study confirm the significant neuroprotection of BASY in the MOG-induced mice model. They could also suggest a novel approach to the treatment of MS-associated disorders. Full article
Show Figures

Figure 1

Figure 1
<p>BASY effectiveness on different parameters of MOG-induced EAE mice. The mice were immunized with MOG 35-33 (emulsified with CFA using a T connector). BASY induced the tolerance against EAE induction in mice: (<b>A</b>) Body weight (BW) of mice (EAE, EAE+BASY and BASY-treated mice). (<b>B</b>) Clinical symptoms recorded post-MOG immunization resulted in gradual increase in paralysis and hind limb inactivity, noted from 11 days of MOG immunization, which became stable after the 18th day post immunization. (<b>C</b>) The total microbial load in pre-administered BASY for 25 days of pre-immunization and MOG-immunized mice after the 16th day. The total bacterial count in fecal samples before and after BASY treatment (Group 1: untreated control group; Group 2: MOG-immunized mice (EAE); Group 3: BASY treated with MOG immunization (EAE+BASY); Group 4: BASY–only treated mice). The uncharacterized colonies were counted using the colony counter and recorded as 10<sup>4</sup> colonies/gram of fecal samples. (<b>D</b>) SCFAs, butyric acid, caproic acid, and propionic acid were quantified using Biovision biochemical kits in the ileal content. The values are measured using a microplate reader and expressed as mM/mg of ileal content. Data were pooled from three independent experiments and shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. (<b>A</b>) microbial load compared using a student <span class="html-italic">t</span>-test. (<b>B</b>) SCFA content was compared using a one-way ANOVA.</p>
Full article ">Figure 2
<p>BASY ameliorated the histopathological hallmarks in the CNS and colon of EAE mice. On the 24th day of MOG post-immunization, spinal cords were euthanized from EAE mice, fixed in 10% formaldehyde following paraffin embedding and sectioned into 4 µm thick sections. (<b>A</b>) Demyelination was assessed in the spinal cords using H&amp;E, and the pathological scores were recorded at 200× magnification power. (<b>B</b>) Infiltration and inflammation scores were noted. (<b>C</b>) Intestinal integrity and mucin damage were assessed using H&amp;E of the proximal colon of EAE mice. (<b>D</b>) Infiltration of T-lymphoid cells, neutrophils, and inflammation scores were noted on the 24th day of MOG post immunization. Data were pooled from three independent experiments and shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Effect of BASY on inflammatory markers (IL-1B, IL-6, IL-17 and TNF-α), oxidative damage (MPO) and cell adhesion (VCAM) markers in CNS of MOG-induced EAE mice. These markers were quantified in MOG-induced EAE mice and BASY-treated mice after 24 days of the experiment. The protein markers were extracted from the CNS of MOG-immunized mice. The tissues were homogenized after 21 days of induction using RIPA lysis buffer. The cytokines and chemokines were quantified using Invitrogen and Cayman ELISA kits. (<b>A</b>) IL-1B was quantified using an Invitrogen kit. The TMB optical variation was quantified at 450 nm by the microplate reader, and values were expressed in pg/mL. (<b>B</b>–<b>D</b>) IL-6, IL-17 and TNF-alpha were quantified using the Cayman kit. (<b>E</b>,<b>F</b>) The oxidative stress marker MPO and adhesion molecule VCAM were quantified in the periphery of the spinal cord tissues, respectively. All data were collected from three individual experiments, pooled, and expressed as mean ± SD (<span class="html-italic">p</span> &lt; 0.05). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 represents significance compared to the MOG vs. MOG+ BASY group. IL—interleukin; TNF-α—tumor necrosis factor alpha; MPO—myeloperoxidase; VCAM—vascular cell adhesion molecule.</p>
Full article ">Figure 4
<p>BASY activates the Treg cell population and the EAE condition by controlling CD4 polarization. (<b>A</b>–<b>D</b>) absolute number of infiltratedCD4, CD8, Treg and FOXP3 positive cells within the CNS of the control, EAE, EAE+BASY, and BASY pretreated MOG-immunized mice groups, respectively. The cells were sorted using the Miltenyi cell separation kit and the total population was counted using the Thermos cell counter. Incidence bars in each graph are expressed as the upper and lower limit of cell population in CNS of EAE and BASY with EAE mice groups. Data were pooled from three independent experiments and shown as mean ± SD. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>BASY augmented the level of neurotransmitters and neuronal hormones in MOG-immunized mice spinal tissues. (<b>A</b>) The peripheral crude lymphocytes were isolated, and total protein was extracted by the rapid freeze method. The melatonin was quantified at two intervals, on the 12th and 24th day of the MOG post-immunization period. The melatonin was quantified using an Abcam hormonal sensitivity kit. (<b>B</b>,<b>C</b>) Acetylcholine, a substrate of esterase, was quantified using the Sigma-Aldrich ELISA kit. Sirtuin protein was quantified using the Invitrogen Mouse ELISA kit. Data were pooled from three independent experiments and shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Peripheral and splenic lymphocytes were regulated by BASY administration in MOG- immunized EAE mice. The differential regulation of lymphocytes in MOG-immunized mice was evaluated by progressive inflammatory cytokine markers using an ELISA kit. CD4 and CD65L lymphocytes were isolated from peripheral lymph nodes of MOG-immunized mice and naïve mice. The isolated cells were restimulated with MOG of 1 µg/mL concentration for 18 h and cells were harvested for cytokine quantification. (<b>A</b>–<b>F</b>) GM-CSF, IFNγ, IL-8, IL-6, IL-17 and TGF-β were quantified in MOG-stimulated peripheral spinal lymph nodes CD4 cells. The isolated cells were re-stimulated with MOG of 1 µg/mL concentration for 12 h and cells were harvested for cytokine quantification. (<b>G</b>–<b>L</b>) GM-CSF, IFNγ, IL-8, IL-6, IL-17 and TGF-β were quantified in MOG-stimulated splenic CD4 cells. CD4 and CD65L lymphocytes were isolated from splenic lymphoid cells (SLC) of MOG -immunized mice and naïve mice. Data were pooled from three independent experiments and shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05. ** <span class="html-italic">p &lt;</span> 0.01. GM -CSF—granulocyte-macrophage colony-stimulating factor; IFN-γ—interferon gamma; IL—interleukin.</p>
Full article ">Figure 7
<p>The cell adhesion molecules regulated by BASY treatment in MOG-induced mice. The spinal cord and colon tissues were homogenized and endothelial cell adhesion molecules VCAM and ICAM were quantified using a protein specific ELISA Kit: (<b>A</b>,<b>B</b>) the differential expression of ICAM and VCAM in the spinal cord after MOG immunization was reduced by BASY treatment; (<b>C</b>,<b>D</b>) the expression and regulation of the invasion of lymphoid cells from the intestinal lamina propria to the peripheral lymph nodes of the spinal cord was quantified. Data were pooled from three independent experiments and shown as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05. ** <span class="html-italic">p</span> &lt; 0.01 (n = 5). ICAM—intracellular adhesion molecule-1; VCAM—vascular cell adhesion molecule-1.</p>
Full article ">Figure 8
<p>Various markers and neurotropic factors regulated by BASY in MOG-induced EAE mice: (<b>A</b>) mRNA expression of MBP, BDNF, and GABA in the spinal cord of MOG-immunized mice. (<b>B</b>) mRNA expression of COX-2 and VEGF in the spinal cord of MOG-immunized mice. (<b>C</b>) MBP, BDNF, GABA, COX-2, and VEGF protein expression was examined by immunoblot and densitometry analysis in the spinal cord and was normalized with laminin and actin as internal controls. (<b>D</b>) mRNA expression of MBP, BDNF, GABA, and COX-2, VEGF in the colon of MOG-immunized mice. (<b>E</b>,<b>F</b>) MBP, BDNF, GABA, COX-2, and VEGF protein expressions were examined by immunoblot and densitometry analysis in the colon and normalized with actin and laminin as internal controls. Results are presented as mean ± SEM (n = 3). When comparing MOG to MOG+BASY groups, * <span class="html-italic">p</span> &lt; 0.05 denotes significance. MBP—myelin basic protein; BDNF—brain-derived neurotrophic factor; GABA—gamma-aminobutyric acid; COX-2—cyclooxygenase-2; and VEGF—vascular endothelial growth factor.</p>
Full article ">
10 pages, 1233 KiB  
Article
Myelin Basic Protein in Oligodendrocyte-Derived Extracellular Vesicles as a Diagnostic and Prognostic Biomarker in Multiple Sclerosis: A Pilot Study
by Cristina Agliardi, Franca Rosa Guerini, Milena Zanzottera, Elisabetta Bolognesi, Silvia Picciolini, Domenico Caputo, Marco Rovaris, Maria Barbara Pasanisi and Mario Clerici
Int. J. Mol. Sci. 2023, 24(1), 894; https://doi.org/10.3390/ijms24010894 - 3 Jan 2023
Cited by 16 | Viewed by 4777
Abstract
Approximately 15% of multiple sclerosis (MS) patients develop a progressive form of disease from onset; this condition (primary progressive-PP) MS is difficult to diagnose and treat, and is associated with a poor prognosis. Extracellular vesicles (EVs) of brain origin isolated from blood and [...] Read more.
Approximately 15% of multiple sclerosis (MS) patients develop a progressive form of disease from onset; this condition (primary progressive-PP) MS is difficult to diagnose and treat, and is associated with a poor prognosis. Extracellular vesicles (EVs) of brain origin isolated from blood and their protein cargoes could function as a biomarker of pathological conditions. We verified whether MBP and MOG content in oligodendrocytes-derived EVs (ODEVs) could be biomarkers of MS and could help in the differential diagnosis of clinical MS phenotypes. A total of 136 individuals (7 clinically isolated syndrome (CIS), 18 PPMS, 49 relapsing remitting (RRMS)) and 70 matched healthy controls (HC) were enrolled. ODEVs were enriched from serum by immune-capture with anti-MOG antibody; MBP and MOG protein cargoes were measured by ELISA. MBP concentration in ODEVs was significantly increased in CIS (p < 0.001), RRMS (p < 0.001) and PPMS (p < 0.001) compared to HC and was correlated with disease severity measured by EDSS and MSSS. Notably, MBP concentration in ODEVs was also significantly augmented in PPMS compared to RRMS (p = 0.004) and CIS (p = 0.03). Logistic regression and ROC analyses confirmed these results. A minimally invasive blood test measuring the concentration of MBP in ODEVs is a promising tool that could facilitate MS diagnosis. Full article
(This article belongs to the Special Issue Molecular Biomarkers in Neurological Diseases)
Show Figures

Figure 1

Figure 1
<p><b>ODEVs characterization.</b> (<b>A</b>): Exo-Check™ Exosome Antibody Array on an exemplificative ODEVs lysate. In the image are visible exosomal associated markers: FLOT1 (flotillin-1), ICAM1 (intercellular adhesion molecule 1), ALIX (programmed cell death 6 interacting protein), CD81 and CD63 (tetraspanins), EpCAM (epithelial cell adhesion molecule), ANXA5 (annexin A5), TSG101 (tumor susceptibility gene 101) and controls (2 positive assay control, negative control: blank and GM130: cis-golgi matrix protein: control for cellular contamination). (<b>B</b>): Immuno-gold (OMGp antigen detected) TEM micrograph of an exemplificative ODEVs preparation. Scale bar: 100 nm. (<b>C</b>): Representative size distribution graph of nanoparticle tracking analysis (NTA) that shows size and concentration of enriched ODEVs in a sample from an RRMS patient; a frame of the video is also shown. Mean ODEVs concentration (particles/mL) ± SD and mean ODEVs diameter (nm) ± SD obtained by NTA analysis from five ODEVs samples from the three conditions (HC, PPMS, and RRMS). ANOVA tests <span class="html-italic">p</span> values are reported.</p>
Full article ">Figure 2
<p><b>MBP in enriched ODEVs in HC, CIS, RR-MS, and PP-MS.</b> (<b>A</b>): Multiple comparison graphs of MBP concentration in enriched ODEVs, respectively in HC, CIS, RRMS, and PPMS subjects; all data are plotted, and median and interquartile range (IQR) are reported. The reported global <span class="html-italic">p</span> values of the differences between the groups of subjects was calculated by Kruskal–Wallis test for non-parametric distributions. <span class="html-italic">p</span> values of post hoc Dwass–Steel–Critchlow–Fligner for pairwise comparisons are also reported. (<b>B</b>): Multiple comparison graphs of MBP concentration in enriched ODEVs, respectively in HC, (CIS + RRMS) and PP-MS subjects; all data are plotted; median and interquartile range (IQR) are reported. The reported global <span class="html-italic">p</span> values of the differences between the groups of subjects was calculated by Kruskal–Wallis test for non-parametric distributions. <span class="html-italic">p</span> values of post hoc Dwass–Steel–Critchlow–Fligner for pairwise comparisons are also reported.</p>
Full article ">Figure 3
<p><b>ROC curve analysis.</b> (<b>A</b>): ROC curves of MBP in enriched ODEVs: HC vs. MS (CIS + RRMS + PPMS). AUC and <span class="html-italic">p</span> value are reported. (<b>B</b>): ROC curves of MBP in enriched ODEVs: PPMS vs. (CIS + RRMS). AUC and <span class="html-italic">p</span> value are reported.</p>
Full article ">Figure 4
<p>(<b>A</b>): <b>Correlation between MBP in ODEVs and clinical scales</b>. Bivariate Pearson’s correlation between MBP concentration in enriched ODEVs and EDSS. (<b>B</b>): Bivariate Pearson’s correlation between MBP concentration in enriched ODEVs and MSSS.</p>
Full article ">
Back to TopTop