Startseite Excessive MALAT1 promotes the immunologic process of neuromyelitis optica spectrum disorder by upregulating BAFF expression
Artikel Open Access

Excessive MALAT1 promotes the immunologic process of neuromyelitis optica spectrum disorder by upregulating BAFF expression

  • Jing Chen , Lijie Zhang , Jingyu Lin , Zeng Wang und Aiyu Lin EMAIL logo
Veröffentlicht/Copyright: 17. Oktober 2023
Veröffentlichen auch Sie bei De Gruyter Brill

Abstract

Increased B cell activating factor (BAFF) expression in patients with neuromyelitis optica spectrum disorder (NMOSD) is associated with B cell overstimulation, but the underlying mechanism remains unclear. This study aimed to reveal the emerging mechanisms that regulate BAFF expression in the inflammatory process of NMOSD. The results showed that the expression of miR-30b-5p was significantly decreased in NMOSD CD14+ monocytes compared with the normal control. Furthermore, we confirmed that metastasis-associated lung adenocarcinoma transcription 1 (MALAT1) is an upstream target of miR-30b-5p, and it could act as a ceRNA and absorb miR-30b-5p with reduced expression of miR-30b-5p. The low expression of miR-30b-5p could not bind to BAFF messenger RNA (mRNA), which resulted in the overexpression of both BAFF mRNA and protein expression. Overexpression of BAFF could bind to the corresponding receptors on B cells, which may initiate activation and proliferation of B cells and increase their production of autoantibodies. Therefore, these findings interpreted that excessive MALAT1 expression in NMOSD mononuclear macrophages led to increased BAFF expression by targeting miR-30b-5p, which caused B cell autoimmune reaction and autoantibodies production, aggravated the disease progression of NMOSD.

Graphical abstract

1 Introduction

Neuromyelitis optica spectrum disorder (NMOSD) is a demyelinating disease of the central nervous system characterized by severe involvement of the optic nerve and the spinal cord, frequent recurrence, and high disability, which tends to onset in middle-aged women [1]. The current evidence suggested that humoral immune abnormalities play an important role in the pathogenesis of NMOSD. Aquaporin4 antibody (AQP4-Ab) has been proved to be specific for NMOSD [2]. In addition, cytokine imbalance is common in patients, and an increase in the concentration of pro-inflammatory cytokines has been reported in the CSF and serum of patients with NMOSD [35]. However, the mechanism of signal crosstalk between cytokines and immune cells in NMOSD is still unknown, which is essential for intervening in the corresponding immune inflammatory damage.

Protein-coding genes have been the focus of research in the past decades, while non-coding RNA have been considered as “garbage” and “noise” generated in the transcription process, but there is increasing evidence that these so-called “by-products” are also involved in cell and tissue growth and homeostasis [6]. MicroRNA has been proven to have a strong regulatory role in many inflammatory reactions or autoimmune diseases [7]. It plays an irreplaceable role in tumors and immunity, and is expected to become diagnostic markers and therapeutic targets of diseases. In the previous study, it was found that miR-30b-5p in the whole blood of NMOSD patients was significantly down-regulated which was statistically significant compared with multiple sclerosis (MS) and the normal control group [8], and was consistent with the results of Keller et al. [9]. Nevertheless, little is known about the role of miR-30b-5p in the biological function of immunocyte and the pathogenesis of autoimmune diseases.

Metastase-associated lung adenocarcinoma transcription 1 (MALAT1), also known as NEAT2, is an 8 KNT long non-coding RNA (IncRNA) located on 11q13 and was originally considered as a poor prognostic parameter for lung cancer patients [10,11]. In recent years, MALAT1 has been found to affect the innate immune response process by affecting the polarization of mononuclear macrophages [12,13]. Meanwhile, emerging evidence supported increase expression of MALAT1 mainly enriched in inflammatory cytokine pathways [14,15]. Therefore, attempts to understand the molecular mechanism of MALAT1 in the mononuclear macrophage cell line of NMOSD is needed for finding its potential pathogenesis.

BAFF is a key survival factor of peripheral blood B cells, and overexpression of BAFF can be found in various autoimmune diseases [16,17]. BAFF secreted by monocytes has been reported to promote pathogenic AQP4 antibody production, which is essential for the development and progression of NMOSD [18]. BAFF is significantly elevated in serum and cerebrospinal fluid of NMOSD patients, and upregulating BAFF is thought to promote and maintain the production of AQP4-Ab [19,20], thereby preventing upstream signaling, then reducing the production of BAFF, which may benefit from the disease progression of NMOSD.

Herein, this study aims to elucidate whether MALAT1/miR-30b-5p/BAFF signal axis is involved in the central nerve injury of NMOSD following immune disorder as well as the underlying mechanisms.

2 Materials and methods

2.1 Participants

Between September 2017 and September 2021, a total of 38 patients were enrolled in this study, all from the First Affiliated Hospital of Fujian Medical University, following 2015 NMOSD diagnostic criteria developed by the international NMO Diagnostic Group (IPND) [21], including 35 patients who were AQP4-IgG positive and three patients were MOG-IgG positive (CBA assay, in house). Clinical data such as duration of disease, frequency of attack, treatment plan, and EDSS score were presented in Table 1. All patients were in the acute stage. Thirty-eight healthy control groups were matched by sex and age. According to recent studies, MOG antibody-associated disease (MOG-AD) has been separated from NMOSD disease as a new disease entity [2224]; therefore, the NMOSD patients referred to in this research only include AQP4 antibody-positive patients. Total transcriptome sequencing was performed in three pairs of untreated NMOSD patients and matched healthy controls. All of the above patients were from the clinical cohort (NCT: 04386018).

Table 1

Clinical and laboratory characteristics of participants in the study

Characteristics Patients (n = 38) Control (n = 38)
Age, mean ± SD year 38.7 ± 11.0 37.4 ± 10.0
Female/male ratio 32/6 35/3
Time since first event (days) 1987.4 ± 1362.3
Recurrence rate (<3/≥3) 26/12
Disease localization
Optic neuritis 6/38
Myelitis 26/38
Brain 14/38
Treatment
Corticosteroids 33/38
Gamma globulin 2/38
Azathioprine 2/38
EDSS score (median[range]) 3 (1–5)
Ratio of AQP4-IgG positivity (±) 35/38
Ratio of MOG-IgG positivity (±) 3/38
Ratio of autoantibody positivity (±) 8/38

2.2 CD14+ cells isolation

From NMOSD patients and controls, 10 mL of blood was collected in vacuette containing ethylenediaminetetraacetic acid and immediately processed according to biosafety rules. Peripheral blood mononuclear cells (PBMCs) were isolated on a Ficoll density gradient (TBDscience, China). All PBMC samples were studied fresh (operative time ≤6 h), which were classified into CD14+ cells bead by positive magnetic separation (Miltenyi Biotec, Germany) according to the manufacturer’s instructions. The sorted cells were determined to have purity greater than 90% by flow cytometry.

2.3 Whole transcriptome sequencing

Three untreated NMOSD patients in the acute stage of the disease and three age–gender matched healthy controls were included. After extracting the total RNA, double-stranded cDNA was synthesized and purified. After terminal repair, a ring connector was connected at both ends of the double-stranded cDNA under the action of DNA ligase, and then the ring was opened by USER enzyme and the second strand of cDNA was disconnected. AMPure XP Beads (Beckman Coulter, Inc.) are used to screen the fragment length to make the target fragment length in the range of 300–400 bp. High-fidelity DNA polymerase is used for PCR amplification and AMPure XP Beads were purified. After quality inspection, the purified library was sequenced by Illumina HiSeq sequencing platform. Clean data were obtained by filtering the sequencing data and compared with the specified reference genome for analysis of transcription/gene expression and differential expression of transcription/gene. The databases involved include Ensemble, GO, KEGG, and STRING. Application software included FastQC, Cutadapt, HISat2, SAMtools, Stringtie, DESeq2, BCFTools, MISA, rMATS, CIRCexplorer, CPAT, R, and Cytoscape.

2.4 Dual-luciferase assay

The promoter validity and miRNA target genes were verified by luciferase double reporter assay. Since miRNA mainly acts on the 3′-UTR of the target gene, the MALAT1 3′-UTR region was constructed behind the reporter gene Luciferase in the vector (Table S1). After comparing the overexpression or interference of miR-30b-5p, changed in reporter gene expression (monitoring changes in luciferase activity) could quantitatively reflect the interaction ability between miR-30b-5p and MALAT1. Combined with site-directed mutation, their action sites were further identified. Plasmid construction: vector: GV272 (Jikai), XbaI/XbaI digestion (Jikai).

HEK293T cells were cultured to logarithmic growth stage, made cell suspension, counted, inoculated in a 24-well culture plate (about 105 cells), and cultured in 5% CO2 incubator at 37°C until the cell fusion degree reached about 60%. The constructed plasmid was transfected with X Tremegene HP (ROCHE) transfection reagent. The expression of fluorescent-labeled genes on the plasmid was observed 24–48 h after transfection to determine the transfection efficiency. Luciferase (Dual-Luciferase® Reporter Assay System, Promega) was detected 48 h after transfection. Firefly luminescence and Renilla luminescence were measured with a microplate tester.

The experimental data in this study were presented as the mean ± standard deviation (SD) of three independent experiments. The statistical significance of the differences in the experimental data was valued by ANOVA followed by Tukey’s post hoc test. The statistical significance level was set as *P < 0.05, **P < 0.01, and ***P < 0.001.

2.5 Quantitative PCR of participant whole blood cells and monocytes

To further confirm the expression levels of miR-30b-5p, MALAT1, and BAFF, total RNA including non-coding RNAs was extracted from the whole blood cells and sorted CD14+ cell of NMOSD patients and matched with the healthy controls by miRNeasy extraction kit (Qiagen, Valenica, CA) using QIAzollysis reagent according to the manufacturer’s instructions. Concentration of RNA was determined using NanoDrop2000 which is very accurate to measure even the small quantities of RNA (Thermo Scientific, USA). Reverse transcription was carried out on extracted RNA in a final volume 20 μL reactions using RT2 first strand kit (Qiagen, Valenica, CA) according to the manufacturer’s instructions. Quantitative PCR was performed with the TaqMan Gene Expression Master Mix (Applied Biosystems) using primers against miR-30b-5p, MALAT1, and BAFF (Table S2). mRNA levels were standardized to the reference human β-actin and GAPDH Endogenous Control (Applied Biosystems). Real-time PCR was done on 20 μL reaction mixture using Rotor geneQ System (Qiagen) with the following conditions: 95°C for 10 min, followed by 45 cycles at 95°C for 15 s and 60°C for 60 s. Gene expression relative to internal control (2−ΔΔCt) was calculated. Fold change was calculated using 2−ΔΔCt for relative quantification. At the same time, we also analyzed the correlation between the three.

2.6 Construction of miR-30b-5p cell lines overexpression and knockdown

THP-1 cells were cultured in medium containing 10% fetal bovine serum at 37°C under 5% CO2, and transfected with plasmid during logarithmic growth phase. THP-1 cells were divided into miR-30b-5p Nor group (transfected with empty vector), miR-30b-5p KD group (transfected with LV-has-mir-30b-5p-inhibition[43448-1]), and miR-30b-5p OE group (transfected with LV-has-mir-30b-5p[41827-3]). The cells were inoculated at a density of 5 × 105/well and cultured in a cell incubator containing 5% CO2 at 37°C. Lipofectamine 2000 (Invitrogen, Carlsbad, CA) was used to transfect cells (4 Ig per well) and the cell adhesion integration rate was 80%. Cells were collected 24 h after transfection, and the expression of miR-30b-5p was detected to determine whether the overexpressed and knockdown miR-30b-5p cell lines were successfully constructed. Quantitative PCR method of miR-30b-5p, MALAT1, and BAFF was the same as the method mentioned above. The difference had statistical significance (P < 0.01).

2.7 Western blot analysis

The above THP-1 cells were lysed in protein lysis buffer containing a proteinase inhibitor (Thermo Fisher Scientific). Lysates were then centrifuged for 15 min at 14,000g at 4°C, and the protein concentration was determined using the Bradford Protein Assay (Thermo Fisher Scientific). Proteins were separated by sodium dodecyl sulfate–polyacrylamide gel electrophoresis using 10% polyacrylamide gels and then transferred onto polyvinylidene difluoride membranes (Millipore, MA, USA). The membranes were blocked with 5% non-fat dry milk in Tris-buffered saline containing 0.1% Tween-20 buffer and immunoblotted with primary antibodies, including anti-BAFF (Abcam) and anti-β-actin (Cell Signaling, BSN, USA). The band intensity was then quantified using Quantity One software (Bio-Rad, CA, USA).

2.8 Statistical analysis

The data were expressed as mean ± SD. Means of two continuous normally distributed variables were compared by independent samples Student’s t-test. Mann–Whitney U-test was used, respectively, to compare means of two and three or more groups of variables not normally distributed. Correlations were analyzed using Pearson’s correlation coefficient. Significance was set as P < 0.05. All analyses were performed using the SPSS 26.0 software and GraphPad Prism 9. A probability (P) value of <0.05 was considered to be significant.

  1. Ethical approval: The research related to human use has been complied with all the relevant national regulations, institutional policies and in accordance with the tenets of the Helsinki Declaration, and has been approved by the authors’ institutional review board or equivalent committee. This study was approved by the ethics commission of the first affiliated hospital of Fujian Medical University.

  2. Informed consent: Informed consent has been obtained from all individuals included in this study.

3 Results

3.1 MALAT1 was aberrantly upregulated expression in patients with NMOSD

In order to search for the upstream regulatory sites of miR-30b-5p, we performed whole transcriptome sequencing based blood in three pairs of patients with NMOSD and matched healthy control. Using the criteria of >2-fold change, P < 0.05, and the threshold of fragments per kilobase of transcript per million (FPKM) mapped reads >50, we identified a total of 247,574 IncRNAs that were differentially expressed between patients and healthy control (Figure 1(a)). The results showed that the expression level of MALAT1 in NMOSD patients was significantly higher than that in healthy controls after difference analysis on IncRNA level (Figure 1(c)).

Figure 1 
                  MALAT1 was overexpressed in NMOSD patients and had binding site with miR-30b-5p. (a) 247574 IncRNAs were differentially expressed in the volcano plot of differentially expressed gene, including MALAT1 (top 10). (b) RNAInter and Starbase v2.0 databases were used to predict the target IncRNA of miR-30b-5p, and MALAT1 was one of them. (c) Columnar statistics of FPKM values of MALAT1 in NMOSD and healthy controls (***P < 0.001). (d) Starbase2.0 database showed that MALAT1 (uplink) had a binding site with miR-30b-5p (downlink). The results of the double luciferase reporter gene experiment showed that miR-30b-5p could bind to the 3′-UTR region of MALAT1 and lost its binding ability after site-directed mutation. In positive control, TRAF6 gene 3′-UTR plasmid was used to overexpress hSA-miR-146b vector plasmid (***P < 0.001).
Figure 1

MALAT1 was overexpressed in NMOSD patients and had binding site with miR-30b-5p. (a) 247574 IncRNAs were differentially expressed in the volcano plot of differentially expressed gene, including MALAT1 (top 10). (b) RNAInter and Starbase v2.0 databases were used to predict the target IncRNA of miR-30b-5p, and MALAT1 was one of them. (c) Columnar statistics of FPKM values of MALAT1 in NMOSD and healthy controls (***P < 0.001). (d) Starbase2.0 database showed that MALAT1 (uplink) had a binding site with miR-30b-5p (downlink). The results of the double luciferase reporter gene experiment showed that miR-30b-5p could bind to the 3′-UTR region of MALAT1 and lost its binding ability after site-directed mutation. In positive control, TRAF6 gene 3′-UTR plasmid was used to overexpress hSA-miR-146b vector plasmid (***P < 0.001).

3.2 MALAT1 was interacted with miR-30b-5p

MALAT1 screened by whole transcriptome sequencing and miR-30b-5p noted in previous studies were predicted in starbase2.0 database and the RNAInter database, and both were found to have binding sites (Figure 1(b)). At the same time, Luciferase reporter gene assay was carried out. The sequence of MALAT1 3′-UTR region was constructed into Luciferase gene of GV272 vector, and then transfected into cells. After overexpression or interference of miR-30b-5p, the expression of Luciferase reporter gene was detected. The interaction between miR-30b-5p and MALAT1 could be quantitatively reflected by using sea kidney luciferase as an internal reference gene. Meanwhile, the sites of action were further identified by site-directed mutation. Experimental results confirmed that MALAT1 could bind to miR-30b-5p and had an interaction relationship (Figure 1(d)).

3.3 Increased accumulation of MALAT1 upregulated BAFF expression by the signaling pathway of miR-30b-5p in the whole blood cells and CD14+ monocytes of NMOSD patients

To further understand the mechanism of action of MALAT1 and miR-30b-5p in the disease process of NMOSD, expression in whole blood cells of 35 NMOSD patients and healthy controls were recruited for preliminary evaluation. Interestingly, MALAT1 and BAFF was overexpressed (Figure 2(a) and (c)), while miR-30b-5p expression was decreased (Figure 2(b)). Combined with functional annotation and signaling pathway prediction by whole transcriptome sequencing, the results showed that the relevant target genes were mainly enriched in pathways similar to systemic lupus erythematosus (SLE) immune inflammation, chronic myeloid cytopathy, and viral infection cancerous lesions (Figure 3(a) and (b)), which was why we focused more on inflammatory alterations in NMOSD myeloid immune cells, and in connection with previous literature reports, CD14+ monocytes became the breakthrough point for the next study.

Figure 2 
                  Expression and relationship of MALAT1, miR-30b-5p, and BAFF in the whole blood cells and CD14+ monocytes of NMOSD patients in qPCR assay. (a–c) 2−△△CT values of MALAT1, miR-30b-5p, and BAFF in the whole blood cells in control group and NMOSD group, respectively (*P < 0.05, ***P < 0.001, ****P < 0.0001). (d–f) 2−△△CT values of MALAT1, miR-30b-5p, and BAFF in CD14+ monocytes in control group and NMOSD group, respectively (*P < 0.05, **P < 0.01, ****P < 0.0001). (g) Correlation between the expression of MALAT1 and miR-30b-5p in CD14+ monocytes. (h) Correlation between the expression of BAFF and miR-30b-5p in CD14+ monocytes.
Figure 2

Expression and relationship of MALAT1, miR-30b-5p, and BAFF in the whole blood cells and CD14+ monocytes of NMOSD patients in qPCR assay. (a–c) 2−△△CT values of MALAT1, miR-30b-5p, and BAFF in the whole blood cells in control group and NMOSD group, respectively (*P < 0.05, ***P < 0.001, ****P < 0.0001). (d–f) 2−△△CT values of MALAT1, miR-30b-5p, and BAFF in CD14+ monocytes in control group and NMOSD group, respectively (*P < 0.05, **P < 0.01, ****P < 0.0001). (g) Correlation between the expression of MALAT1 and miR-30b-5p in CD14+ monocytes. (h) Correlation between the expression of BAFF and miR-30b-5p in CD14+ monocytes.

Figure 3 
                  Bioinformatic analysis of whole transcriptome sequencing, then quantitative PCR assay and western blot analysis for BAFF in miR-30b-5p OE group, miR-30b-5p KD group, and miR-30b-5p Nor group. (a) KEGG analysis and (b) GO analysis. (c) The respective bar graphs showed that the expression of MALAT1 gene was increased in miR-30b-5p KD group, and the expression of MALAT1 gene was decreased in miR-30b-5p OE group (**P < 0.01, ****P < 0.0001). (d) The respective bar graphs showed that the expression of BAFF gene was increased in miR-30b-5p KD group, and the expression of BAFF gene was decreased in miR-30b-5p OE group (**P < 0.01, ***P < 0.001, ****P < 0.0001). The cellular status after transfection with LV-has-miR-30b-5p-inhibition[43448-1] and LV-has-miR-30b-5p[41827-3] for 72 h. (e) Representative western blot for BAFF among three group (miR-30b-5p Nor, miR-30b-5p KD, and miR-30b-5p OE).
Figure 3

Bioinformatic analysis of whole transcriptome sequencing, then quantitative PCR assay and western blot analysis for BAFF in miR-30b-5p OE group, miR-30b-5p KD group, and miR-30b-5p Nor group. (a) KEGG analysis and (b) GO analysis. (c) The respective bar graphs showed that the expression of MALAT1 gene was increased in miR-30b-5p KD group, and the expression of MALAT1 gene was decreased in miR-30b-5p OE group (**P < 0.01, ****P < 0.0001). (d) The respective bar graphs showed that the expression of BAFF gene was increased in miR-30b-5p KD group, and the expression of BAFF gene was decreased in miR-30b-5p OE group (**P < 0.01, ***P < 0.001, ****P < 0.0001). The cellular status after transfection with LV-has-miR-30b-5p-inhibition[43448-1] and LV-has-miR-30b-5p[41827-3] for 72 h. (e) Representative western blot for BAFF among three group (miR-30b-5p Nor, miR-30b-5p KD, and miR-30b-5p OE).

The relationship between MALAT1 and miR-30b-5p was extended to monocyte validation in patients, then we studied whether BAFF was identified as a downstream molecular target of both miR-30b-5p. Real time PCR results showed that MALAT1 and BAFF were significantly increased in CD14+ cells of NMOSD patients (Figure 2(d) and (f)), while miR-30b-5p was significantly decreased (Figure 2(e)). Meanwhile, the expression of MALAT1 was negatively correlated with miR-30b-5p (r = −0.41, P = 0.02, Figure 2(g)), and the expression of BAFF was negatively correlated with miR-30b-5p (r = −0.68, P = 0.0001, Figure 2(h)). Experimental evidence suggested MALAT1/miR-30b-5p/BAFF signal axis participated in immunity process of NMOSD monocyte. In this experiment, the purity of CD14+ cells sorted by flow-verified magnetic beads can reach over 90% (Figure S1).

In addition, comparing NMOSD patients, in CD14+ monocytes from three MOG-AD patients, MALAT1 expression was reduced and miR-30b-5p expression was elevated, while BAFF mRNA overexpression was not significant (P = 0.7459, Figure S2).

3.4 MALAT1 bound to miR-30b-5p to upregulate BAFF expression and trigger immune inflammatory responses

THP-1 was transfected with different plasmids to construct three cell lines: overexpression of miR-30b-5p THP-1 cell group (miR-30b-5p OE), knockdown of miR-30b-5p THP-1 cell group (miR-30b-5p KD), and empty viral THP-1 cells (miR-30b-5p Nor). The correlation between miR-30b-5p and BAFF was further verified in the above three types of cells. At the RNA level, real time PCR detection showed that the expression of MALAT1 was upregulated in THP-1 cells down-regulated miR-30b-5p, while the expression of MALAT1 was down-regulated in THP-1 cells overexpressing miR-30b-5p (Figure 3(c)). In addition, BAFFmRNA expression was up-regulated in THP-1 cells that down-regulated miR-30b-5p and down-regulated in THP-1 cells that overexpressed miR-30b-5p (Figure 3(d)). At the protein level, down-regulation of BAFF protein expression was found in THP-1 cells that overexpressed miR-30b-5p, while the down-regulation of BAFF protein expression was found in THP-1 cells that knocked down miR-30b-5p (Figure 3(e)).

4 Discussion

Monocytes have received increasing attention for their role in exacerbating inflammatory storms in autoimmune diseases such as MS, SLE, and rheumatoid arthritis [2527]. Currently, NMOSD is believed to be mainly due to B cell-mediated autoimmune diseases, but monocytes and other innate immune cells also play an indispensable role [28,29], and there are still gaps in the molecular mechanism of monocytes’ involvement in humoral immunity of NMOSD. In a previous research, nine candidate miRNAs were isolated for real-time PCR from whole blood from 20 healthy controls, 45 NMOSD, and 17 MS patients. Among the mircoRNA, we found that miR-30b-5p was significantly decreased by about 1/2 in NMOSD patients compared with the control group, and was negatively correlated with the degree of intracranial lesions in NMOSD [8], and Keller et al. found similar change [9].

Past studies have found a microarray analysis of mRNA and IncRNA in peripheral mononuclear cells of patients with NMOSD, and found that there were numerous epigenetic differences in patients with NMOSD compared to healthy people, suggesting that these factors would be key to early diagnosis, appropriate treatment, and better prognosis of patients with NMOSD [30]. By targeting mircoRNA, these lncRNAs play an important role in guiding the development of a variety of immune cells and in controlling the dynamic transcription program of immune cell activation markers [31,32]. In order to understand the molecular mechanism of miR-30b-5p in NMOSD disease, we identified the upstream target of MALAT1 through total transcriptomic data and software prediction results. MALAT1 could act as the ceRNA of miR-30b-5p, thereby inhibiting the expression of miR-30b-5p. Most of the previous studies on MALAT1 were mainly on lung cancer and other tumors [33,34]. Our results refreshed the understanding of MALAT1 in the autoimmune process, and could also explain the corresponding molecular basis of MALAT1 in tumor immune tolerance escape [35,36].

There is a genetic susceptibility to autoimmune diseases, a single-cell RNA sequencing study suggested experimental evidence of increased inflammatory cytokine gene expression in immune cells during the early immune response of NMOSD patients [37], and We were more concerned with the risk of immune-inflammatory predisposition brought by differently expressed genetic information, which was reflected not only in the production of pro-inflammatory cytokines, but also in the signal crosstalk between inflammatory cells brought by the cytokines [38]. What were the downstream immunoinflammatory targets of miR-30b-5p? In a study of Sjogren’s syndrome, miR-30b-5p could negatively regulate the expression of BAFF in monocytes [39]. This caught our attention. Therefore, in this study, we hypothesized that NMOSD miR-30b-5p could also negatively regulate BAFF, while down-regulated miR-30b-5p reversed this process, leading to upregulation of BAFF expression. CD14+ monocyte levels in a large number of patients also confirmed our idea. In order to further determine the relationship among the axis of MALAT1–miR-30b-5p–BAFF, the THP-1 cell lines of miR-30b-5p overexpression and knockdown were constructed to validate this potential signaling axis, which sheds light on the future of targeted therapy for NMOSD.

As a mimic of NMOSD disease, a similar correspondence of the MALAT1/miR-30b-5p/BAFF signaling axis does not seem to be found in MOG-AD patients, and we hypothesize that this pathway may occur only in NMOSD patients, which promises to be a means of differential diagnosis between the two. Multiple studies have suggested that the NMOSD lesions were more severely involved than those of MOG-AD [4042], and a recent pathologic research has shown that immune-inflammatory cell infiltration in the acute phase of MOG-IgG experimental autoimmune encephalomyelitis (EAE) mice was less common than in AQP4-IgG EAE, and monocyte macrophage infiltration was not seen, leading to the early onset of inflammation in NMOSD [43], which fitted with our findings that the level of expression of BAFF mRNA, an inflammatory cytokine secreted by monocytes, was slightly lower in MOG-AD patients than in NMOSD patients, and that, because this study enrolled fewer MOG-AD patients, resulting in a slightly weaker level of evidence, and the sample size could be expanded later to clarify the specific regulatory mechanisms involved in the variability.

BAFF, a member of the tumor necrosis factor family that plays an important role in the activation of B cells, was first discovered around 2000 [44,45]. It is mainly divided into two forms: membrane-bound BAFF and soluble BAFF [46,47]. Currently, there are many research studies on soluble BAFF, but the understanding of membrane-bound BAFF is still not deep enough [16,48]. Our results showed that in the disease process of NMOSD, down-regulation of miR-30b-5p led to increased expression of membrane-bound BAFF, which in turn triggered the secretion of soluble BAFF during protein hydrolysis and affected the immune response process.

This study has two limitations. First, MALAT1 mainly increased the expression of membrane-bound BAFF, but whether it increased the expression of soluble BAFF remains unknown. The molecular mechanism behind it deserves further exploration. Second, more cytological and zoological experimental evidence is needed to confirm whether the downstream effect on the activation and proliferation of B cells in the relapse stage of NMOSD disease is related to the upregulated membrane-bound BAFF content of monocytes.

5 Conclusion

In brief, this study suggested that overexpression of MALAT1 could bind and absorb miR-30b-5p in NMOSD monocytes, and then miR-30b-5p reduced the inhibition of BAFF transcription translation, resulting in overexpression of the corresponding receptors of BAFF binding B cells, which may trigger a series of activated B cell proliferation responses. These findings elucidated a novel molecular mechanism by which MALAT1 mediated indirect B cell activation. In addition, it provided an explanation for the over-activation of NMOSD B cells caused by miR-30b-5p down-regulation.


# These authors have contributed equally to this work and share first authorship.


Acknowledgements

The authors thank their patients for participating in this study and the clinical neuroimmunology team for recruiting the patients.

  1. Funding information: This work was supported by the Fujian Province Natural Science Foundation Research Project (grant number 2019J01311) (A.L.), the Fujian Province Science and Technology Innovation Joint Funding Project (grant number 2020Y9128) (A.L.), and the National Natural Science Foundation of China (grants number 82271375) (A.L.).

  2. Author contributions: Jing Chen: formal analysis (lead), investigation (lead), methodology (lead), software (lead), validation (equal), writing – orginal draft (equal), writing – review and editing (supporting). Lijie Zhang: formal analysis (lead), investigation (lead), methodology (lead), software (lead), validation (equal), writing – orginal draft (lead), writing – review and editing (equal). Jingyu Lin: formal analysis (supporting), investigation (supporting), methodology (supporting), software (supporting). Zeng Wang: formal analysis (equal), investigation (supporting), methodology (supporting), software (supporting). Aiyu Lin: conceptualization (lead), funding acquisition (lead), supervision (lead), writing – review and editing (lead).

  3. Conflict of interest: Authors state no conflict of interest.

  4. Data availability statement: The datasets generated during and/or analyzed during the current study are available from the corresponding author on reasonable request.

References

[1] Jarius S, Paul F, Weinshenker BG, Levy M, Kim HJ, Wildemann B. Neuromyelitis optica. Nat Rev Dis Primers. 2020;6(1):85.10.1038/s41572-020-0214-9Suche in Google Scholar PubMed

[2] Lennon VA, Wingerchuk DM, Kryzer TJ, Pittock SJ, Lucchinetti CF, Fujihara K, et al. A serum autoantibody marker of neuromyelitis optica: distinction from multiple sclerosis. Lancet. 2004;364(9451):2106–12.10.1016/S0140-6736(04)17551-XSuche in Google Scholar PubMed

[3] Wang Y, Zhou Y, Sun X, Lu T, Wei L, Fang L, et al. Cytokine and chemokine profiles in patients with neuromyelitis optica spectrum disorder. Neuroimmunomodulation. 2016;23(5–6):352–8.10.1159/000464135Suche in Google Scholar PubMed

[4] Yandamuri SS, Jiang R, Sharma A, Cotzomi E, Zografou C, Ma AK, et al. High-throughput investigation of molecular and cellular biomarkers in NMOSD. Neurol Neuroimmunol Neuroinflamm. 2020;7(5):e852.10.1212/NXI.0000000000000852Suche in Google Scholar PubMed PubMed Central

[5] Fujihara K, Bennett JL, de Seze J, Haramura M, Kleiter I, Weinshenker BG, et al. Interleukin-6 in neuromyelitis optica spectrum disorder pathophysiology. Neurol Neuroimmunol Neuroinflamm. 2020;7(5):e841.10.1212/NXI.0000000000000841Suche in Google Scholar PubMed PubMed Central

[6] Wright MW, Bruford EA. Naming ‘junk’: human non-protein coding RNA (ncRNA) gene nomenclature. Hum Genomics. 2011;5(2):90–8.10.1186/1479-7364-5-2-90Suche in Google Scholar PubMed PubMed Central

[7] Garo LP, Murugaiyan G. Contribution of MicroRNAs to autoimmune diseases. Cell Mol Life Sci. 2016;73(10):2041–51.10.1007/s00018-016-2167-4Suche in Google Scholar PubMed

[8] Chen J, Zhu J, Wang Z, Yao X, Wu X, Liu F, et al. MicroRNAs correlate with multiple sclerosis and neuromyelitis optica spectrum disorder in a chinese population. Med Sci Monit. 2017;23:2565–83.10.12659/MSM.904642Suche in Google Scholar

[9] Keller A, Leidinger P, Meese E, Haas J, Backes C, Rasche L, et al. Next-generation sequencing identifies altered whole blood microRNAs in neuromyelitis optica spectrum disorder which may permit discrimination from multiple sclerosis. J Neuroinflammation. 2015;12:196.10.1186/s12974-015-0418-1Suche in Google Scholar PubMed PubMed Central

[10] Ji P, Diederichs S, Wang W, Böing S, Metzger R, Schneider PM, et al. MALAT-1, a novel noncoding RNA, and thymosin beta4 predict metastasis and survival in early-stage non-small cell lung cancer. Oncogene. 2003;22(39):8031–41.10.1038/sj.onc.1206928Suche in Google Scholar PubMed

[11] Schmidt LH, Spieker T, Koschmieder S, Schäffers S, Humberg J, Jungen D, et al. The long noncoding MALAT-1 RNA indicates a poor prognosis in non-small cell lung cancer and induces migration and tumor growth. J Thorac Oncol. 2011;6(12):1984–92.10.1097/JTO.0b013e3182307eacSuche in Google Scholar PubMed

[12] Gast M, Rauch BH, Nakagawa S, Haghikia A, Jasina A, Haas J, et al. Immune system-mediated atherosclerosis caused by deficiency of long non-coding RNA MALAT1 in ApoE-/-mice. Cardiovasc Res. 2019;115(2):302–14.10.1093/cvr/cvy202Suche in Google Scholar PubMed

[13] Liu W, Wang Z, Liu L, Yang Z, Liu S, Ma Z, et al. LncRNA Malat1 inhibition of TDP43 cleavage suppresses IRF3-initiated antiviral innate immunity. Proc Natl Acad Sci U S A. 2020;117(38):23695–706.10.1073/pnas.2003932117Suche in Google Scholar PubMed PubMed Central

[14] Tang R, Meng T, Lin W, Shen C, Ooi JD, Eggenhuizen PJ, et al. A partial picture of the single-cell transcriptomics of human IgA nephropathy. Front Immunol. 2021;12:645988.10.3389/fimmu.2021.645988Suche in Google Scholar PubMed PubMed Central

[15] Yao MY, Zhang WH, Ma WT, Liu QH, Xing LH, Zhao GF. Long non-coding RNA MALAT1 exacerbates acute respiratory distress syndrome by upregulating ICAM-1 expression via microRNA-150-5p downregulation. Aging (Albany NY). 2020;12(8):6570–85.10.18632/aging.102953Suche in Google Scholar PubMed PubMed Central

[16] Eslami M, Schneider P. Function, occurrence and inhibition of different forms of BAFF. Curr Opin Immunol. 2021;71:75–80.10.1016/j.coi.2021.06.009Suche in Google Scholar PubMed

[17] Mackay F, Schneider P. Cracking the BAFF code. Nat Rev Immunol. 2009;9(7):491–502.10.1038/nri2572Suche in Google Scholar PubMed

[18] Wilson R, Makuch M, Kienzler AK, Varley J, Taylor J, Woodhall M, et al. Condition-dependent generation of aquaporin-4 antibodies from circulating B cells in neuromyelitis optica. Brain. 2018;141(4):1063–74.10.1093/brain/awy010Suche in Google Scholar PubMed PubMed Central

[19] Okada K, Matsushita T, Kira J, Tsuji S. B-cell activating factor of the TNF family is upregulated in neuromyelitis optica. Neurology. 2010;74(2):177–8.10.1212/WNL.0b013e3181c919eeSuche in Google Scholar PubMed

[20] Wang H, Wang K, Zhong X, Qiu W, Dai Y, Wu A, et al. Cerebrospinal fluid BAFF and APRIL levels in neuromyelitis optica and multiple sclerosis patients during relapse. J Clin Immunol. 2012;32(5):1007–11.10.1007/s10875-012-9709-9Suche in Google Scholar PubMed

[21] Wingerchuk DM, Banwell B, Bennett JL, Cabre P, Carroll W, Chitnis T, et al. International consensus diagnostic criteria for neuromyelitis optica spectrum disorders. Neurology. 2015;85(2):177–89.10.1212/WNL.0000000000001729Suche in Google Scholar PubMed PubMed Central

[22] Jurynczyk M, Messina S, Woodhall MR, Raza N, Everett R, Roca-Fernandez A, et al. Clinical presentation and prognosis in MOG-antibody disease: a UK study. Brain. 2017;140(12):3128–38.10.1093/brain/awx276Suche in Google Scholar PubMed

[23] Jarius S, Paul F, Aktas O, Asgari N, Dale RC, de Seze J, et al. MOG encephalomyelitis: international recommendations on diagnosis and antibody testing. J Neuroinflammation. 2018;15(1):134.10.1186/s12974-018-1144-2Suche in Google Scholar PubMed PubMed Central

[24] Banwell B, Bennett JL, Marignier R, Kim HJ, Brilot F, Flanagan EP, et al. Diagnosis of myelin oligodendrocyte glycoprotein antibody-associated disease: International MOGAD Panel proposed criteria. Lancet Neurol. 2023;22(3):268–82.10.1016/S1474-4422(22)00431-8Suche in Google Scholar PubMed

[25] Radandish M, Khalilian P, Esmaeil N. The role of distinct subsets of macrophages in the pathogenesis of MS and the impact of different therapeutic agents on these populations. Front Immunol. 2021;12:667705.10.3389/fimmu.2021.667705Suche in Google Scholar PubMed PubMed Central

[26] Hirose S, Lin Q, Ohtsuji M, Nishimura H, Verbeek JS. Monocyte subsets involved in the development of systemic lupus erythematosus and rheumatoid arthritis. Int Immunol. 2019;31(11):687–96.10.1093/intimm/dxz036Suche in Google Scholar PubMed PubMed Central

[27] Morell M, Varela N, Marañón C. Myeloid populations in systemic autoimmune diseases. Clin Rev Allergy Immunol. 2017;53(2):198–218.10.1007/s12016-017-8606-7Suche in Google Scholar PubMed

[28] Kong BS, Kim Y, Kim GY, Hyun JW, Kim SH, Jeong A, et al. Increased frequency of IL-6-producing non-classical monocytes in neuromyelitis optica spectrum disorder. J Neuroinflammation. 2017;14(1):191.10.1186/s12974-017-0961-zSuche in Google Scholar PubMed PubMed Central

[29] Gelfand JM, Cotter J, Klingman J, Huang EJ, Cree BA. Massive CNS monocytic infiltration at autopsy in an alemtuzumab-treated patient with NMO. Neurol Neuroimmunol Neuroinflamm. 2014;1(3):e34.10.1212/NXI.0000000000000034Suche in Google Scholar PubMed PubMed Central

[30] Xu J, Zhang F, Gao C, Ma X, Peng X, Kong D, et al. Microarray analysis of lncRNA and mRNA expression profiles in patients with neuromyelitis optica. Mol Neurobiol. 2017;54(3):2201–8.10.1007/s12035-016-9754-0Suche in Google Scholar PubMed PubMed Central

[31] Atianand MK, Caffrey DR, Fitzgerald KA. Immunobiology of long noncoding RNAs. Annu Rev Immunol. 2017;35:177–98.10.1146/annurev-immunol-041015-055459Suche in Google Scholar PubMed PubMed Central

[32] Mowel WK, McCright SJ, Kotzin JJ, Collet MA, Uyar A, Chen X, et al. Group 1 innate lymphoid cell lineage identity is determined by a cis-regulatory element marked by a long non-coding RNA. Immunity. 2017;47(3):435–49.e8.10.1016/j.immuni.2017.08.012Suche in Google Scholar PubMed PubMed Central

[33] Ahmad S, Abbas M, Ullah MF, Aziz MH, Beylerli O, Alam MA, et al. Long non-coding RNAs regulated NF-κB signaling in cancer metastasis: micromanaging by not so small non-coding RNAs. Semin Cancer Biol. 2022;85:155–63.10.1016/j.semcancer.2021.07.015Suche in Google Scholar PubMed

[34] Liu SJ, Dang HX, Lim DA, Feng FY, Maher CA. Long noncoding RNAs in cancer metastasis. Nat Rev Cancer. 2021;21(7):446–60.10.1038/s41568-021-00353-1Suche in Google Scholar PubMed PubMed Central

[35] Wang QM, Lian GY, Song Y, Huang YF, Gong Y. LncRNA MALAT1 promotes tumorigenesis and immune escape of diffuse large B cell lymphoma by sponging miR-195. Life Sci. 2019;231:116335.10.1016/j.lfs.2019.03.040Suche in Google Scholar PubMed

[36] Agarwal S, Vierbuchen T, Ghosh S, Chan J, Jiang Z, Kandasamy RK, et al. The long non-coding RNA LUCAT1 is a negative feedback regulator of interferon responses in humans. Nat Commun. 2020;11(1):6348.10.1038/s41467-020-20165-5Suche in Google Scholar PubMed PubMed Central

[37] Zhang C, Zhang TX, Liu Y, Jia D, Zeng P, Du C, et al. B-Cell compartmental features and molecular basis for therapy in autoimmune disease. Neurol Neuroimmunol Neuroinflamm. 2021;8(6):e1070.10.1212/NXI.0000000000001070Suche in Google Scholar PubMed PubMed Central

[38] Steri M, Orrù V, Idda ML, Pitzalis M, Pala M, Zara I, et al. Overexpression of the cytokine BAFF and autoimmunity risk. N Engl J Med. 2017;376(17):1615–26.10.1056/NEJMoa1610528Suche in Google Scholar PubMed PubMed Central

[39] Wang-Renault SF, Boudaoud S, Nocturne G, Roche E, Sigrist N, Daviaud C, et al. Deregulation of microRNA expression in purified T and B lymphocytes from patients with primary Sjögren's syndrome. Ann Rheum Dis. 2018;77(1):133–40.10.1136/annrheumdis-2017-211417Suche in Google Scholar PubMed PubMed Central

[40] Sotirchos ES, Filippatou A, Fitzgerald KC, Salama S, Pardo S, Wang J, et al. Aquaporin-4 IgG seropositivity is associated with worse visual outcomes after optic neuritis than MOG-IgG seropositivity and multiple sclerosis, independent of macular ganglion cell layer thinning. Mult Scler. 2020;26(11):1360–71.10.1177/1352458519864928Suche in Google Scholar PubMed PubMed Central

[41] Pache F, Zimmermann H, Mikolajczak J, Schumacher S, Lacheta A, Oertel FC, et al. MOG-IgG in NMO and related disorders: a multicenter study of 50 patients. Part 4: afferent visual system damage after optic neuritis in MOG-IgG-seropositive versus AQP4-IgG-seropositive patients. J Neuroinflammation. 2016;13(1):282.10.1186/s12974-016-0720-6Suche in Google Scholar PubMed PubMed Central

[42] Sato DK, Callegaro D, Lana-Peixoto MA, Waters PJ, de Haidar Jorge FM, Takahashi T, et al. Distinction between MOG antibody-positive and AQP4 antibody-positive NMO spectrum disorders. Neurology. 2014;82(6):474–81.10.1212/WNL.0000000000000101Suche in Google Scholar PubMed PubMed Central

[43] Remlinger J, Bagnoud M, Meli I, Massy M, Hoepner R, Linington C, et al. Modeling MOG antibody-associated disorder and neuromyelitis optica spectrum disorder in animal models: visual system manifestations. Neurol Neuroimmunol Neuroinflamm. 2023;10(5):e200141.10.1212/NXI.0000000000200141Suche in Google Scholar PubMed

[44] Mackay F, Woodcock SA, Lawton P, Ambrose C, Baetscher M, Schneider P, et al. Mice transgenic for BAFF develop lymphocytic disorders along with autoimmune manifestations. J Exp Med. 1999;190(11):1697–710.10.1084/jem.190.11.1697Suche in Google Scholar PubMed PubMed Central

[45] Karpusas M, Cachero TG, Qian F, Boriack-Sjodin A, Mullen C, Strauch K, et al. Crystal structure of extracellular human BAFF, a TNF family member that stimulates B lymphocytes. J Mol Biol. 2002;315(5):1145–54.10.1006/jmbi.2001.5296Suche in Google Scholar PubMed

[46] Liu Y, Hong X, Kappler J, Jiang L, Zhang R, Xu L, et al. Ligand–receptor binding revealed by the TNF family member TALL-1. Nature. 2003;423(6935):49–56.10.1038/nature01543Suche in Google Scholar PubMed

[47] Bossen C, Schneider P. BAFF, APRIL and their receptors: structure, function and signaling. Semin Immunol. 2006;18(5):263–75.10.1016/j.smim.2006.04.006Suche in Google Scholar PubMed

[48] Lee SM, Kim WJ, Suk K, Lee WH. Cell to cell interaction can activate membrane-bound APRIL which are expressed on inflammatory macrophages. Immune Netw. 2010;10(5):173–80.10.4110/in.2010.10.5.173Suche in Google Scholar PubMed PubMed Central

Received: 2023-04-02
Revised: 2023-08-13
Accepted: 2023-08-16
Published Online: 2023-10-17

© 2023 the author(s), published by De Gruyter

This work is licensed under the Creative Commons Attribution 4.0 International License.

Artikel in diesem Heft

  1. Research Articles
  2. HIF-1α participates in secondary brain injury through regulating neuroinflammation
  3. Omega-3 polyunsaturated fatty acids alleviate early brain injury after traumatic brain injury by inhibiting neuroinflammation and necroptosis
  4. The correlation between non-arteritic anterior ischemic optic neuropathy and cerebral infarction
  5. Enriched environment can reverse chronic sleep deprivation-induced damage to cellular plasticity in the dentate gyrus of the hippocampus
  6. Middle cerebral artery dynamic cerebral autoregulation is impaired by infarctions in the anterior but not the posterior cerebral artery territory in patients with mild strokes
  7. Leptin ameliorates Aβ1-42-induced Alzheimer’s disease by suppressing inflammation via activating p-Akt signaling pathway
  8. TIPE2 knockdown exacerbates isoflurane-induced postoperative cognitive impairment in mice by inducing activation of STAT3 and NF-κB signaling pathways
  9. Does the patellar tendon reflex affect the postural stability in stroke patients with blocked vision?
  10. Inactivation of CACNA1H induces cell apoptosis by initiating endoplasmic reticulum stress in glioma
  11. miR-101-3p improves neuronal morphology and attenuates neuronal apoptosis in ischemic stroke in young mice by downregulating HDAC9
  12. A custom-made weight-drop impactor to produce consistent spinal cord injury outcomes in a rat model
  13. Arterial spin labeling for moyamoya angiopathy: A preoperative and postoperative evaluation method
  14. Thyroid hormone levels paradox in acute ischemic stroke
  15. Geniposide protected against cerebral ischemic injury through the anti-inflammatory effect via the NF-κB signaling pathway
  16. The clinical characteristics of acute cerebral infarction patients with thalassemia in a tropic area in China
  17. Comprehensive behavioral study of C57BL/6.KOR-ApoEshl mice
  18. Incomplete circle of Willis as a risk factor for intraoperative ischemic events during carotid endarterectomies performed under regional anesthesia – A prospective case-series
  19. HOTAIRM1 knockdown reduces MPP+-induced oxidative stress injury of SH-SY5Y cells by activating the Nrf2/HO-1 pathway
  20. Esmolol inhibits cognitive impairment and neuronal inflammation in mice with sepsis-induced brain injury
  21. EHMT2 affects microglia polarization and aggravates neuronal damage and inflammatory response via regulating HMOX1
  22. Hematoma evacuation based on active strategies versus conservative treatment in the management of moderate basal ganglia hemorrhage: A retrospective study
  23. Knockdown of circEXOC6 inhibits cell progression and glycolysis by sponging miR-433-3p and mediating FZD6 in glioma
  24. CircYIPF6 regulates glioma cell proliferation, apoptosis, and glycolysis through targeting miR-760 to modulate PTBP1 expression
  25. Relationship between serum HIF-1α and VEGF levels and prognosis in patients with acute cerebral infarction combined with cerebral-cardiac syndrome
  26. The promoting effect of modified Dioscorea pills on vascular remodeling in chronic cerebral hypoperfusion via the Ang/Tie signaling pathway
  27. Effects of enriched environment on the expression of β-amyloid and transport-related proteins LRP1 and RAGE in chronic sleep-deprived mice
  28. An interventional study of baicalin on neuronal pentraxin-1, neuronal pentraxin-2, and C-reactive protein in Alzheimer’s disease rat model
  29. PD98059 protects SH-SY5Y cells against oxidative stress in oxygen–glucose deprivation/reperfusion
  30. TPVB and general anesthesia affects postoperative functional recovery in elderly patients with thoracoscopic pulmonary resections based on ERAS pathway
  31. Brain functional connectivity and network characteristics changes after vagus nerve stimulation in patients with refractory epilepsy
  32. Association between RS3763040 polymorphism of the AQP4 and idiopathic intracranial hypertension in a Spanish Caucasian population
  33. Effects of γ-oryzanol on motor function in a spinal cord injury model
  34. Electroacupuncture inhibits the expression of HMGB1/RAGE and alleviates injury to the primary motor cortex in rats with cerebral ischemia
  35. Effects of edaravone dexborneol on neurological function and serum inflammatory factor levels in patients with acute anterior circulation large vessel occlusion stroke
  36. CST3 alleviates bilirubin-induced neurocytes’ damage by promoting autophagy
  37. Excessive MALAT1 promotes the immunologic process of neuromyelitis optica spectrum disorder by upregulating BAFF expression
  38. Evaluation of cholinergic enzymes and selected biochemical parameters in the serum of patients with a diagnosis of acute subarachnoid hemorrhage
  39. 7-Day National Institutes of Health Stroke Scale as a surrogate marker predicting ischemic stroke patients’ outcome following endovascular therapy
  40. Cdk5 activation promotes Cos-7 cells transition towards neuronal-like cells
  41. 10.1515/tnsci-2022-0313
  42. PPARα agonist fenofibrate prevents postoperative cognitive dysfunction by enhancing fatty acid oxidation in mice
  43. Predicting functional outcome in acute ischemic stroke patients after endovascular treatment by machine learning
  44. EGCG promotes the sensory function recovery in rats after dorsal root crush injury by upregulating KAT6A and inhibiting pyroptosis
  45. Preoperatively administered single dose of dexketoprofen decreases pain intensity on the first 5 days after craniotomy: A single-centre placebo-controlled, randomized trial
  46. Myeloarchitectonic maps of the human cerebral cortex registered to surface and sections of a standard atlas brain
  47. The BET inhibitor apabetalone decreases neuroendothelial proinflammatory activation in vitro and in a mouse model of systemic inflammation
  48. Carthamin yellow attenuates brain injury in a neonatal rat model of ischemic–hypoxic encephalopathy by inhibiting neuronal ferroptosis in the hippocampus
  49. Functional connectivity in ADHD children doing Go/No-Go tasks: An fMRI systematic review and meta-analysis
  50. Review Articles
  51. Human prion diseases and the prion protein – what is the current state of knowledge?
  52. Nanopharmacology as a new approach to treat neuroinflammatory disorders
  53. Case Report
  54. Deletion as novel variants in VPS13B gene in Cohen syndrome: Case series
  55. Commentary
  56. Translation of surface electromyography to clinical and motor rehabilitation applications: The need for new clinical figures
  57. Revealing key role of T cells in neurodegenerative diseases, with potential to develop new targeted therapies
  58. Retraction
  59. Retraction of “Eriodictyol corrects functional recovery and myelin loss in SCI rats”
  60. Special Issue “Advances in multimedia-based emerging technologies...”
  61. Evaluation of the improvement of walking ability in patients with spinal cord injury using lower limb rehabilitation robots based on data science
Heruntergeladen am 5.9.2025 von https://www.degruyterbrill.com/document/doi/10.1515/tnsci-2022-0306/html?srsltid=AfmBOoqc25-flI5604l1fn64mjoytoJt-pxDT1x_WTYXyWXjBgY5O4W3
Button zum nach oben scrollen