1 Introduction
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease that affects upper and lower motor neurons, normally culminating in respiratory failure and death within 5 years of disease onset [
1–
4]. Despite recent expansion of the disease-associated gene list, the etiology of ALS remains largely unclear in sporadic patients without known genetic causes [
5–
7]. Several common ALS mutant genes, such as
C9orf72 [
8],
TARDBP [
9], and
SOD1 [
10], are notably implicated in aberrant immune responses of the central nervous system (CNS), and mutations in
OPTN,
TBK1, and
TNIP1 can similarly compromise immune function and induce neuroinflammation [
11]. However, the complex interplay between genetics and dysregulated inflammatory pathways in ALS is not fully understood, partly due to a lack of evidence that ALS is primarily an immunogenic neurodegenerative disorder because immunoregulatory drugs have so far failed to impede disease progression in most clinical trials [
12,
13]. Therefore, investigating the potential role of immune dysregulation in the disease is crucial to advance the understanding of the intricate mechanisms underlying ALS pathogenesis, possibly necessitating the identification and characterization of novel regulatory mechanisms.
The N
6-methyladenosine (m
6A) modification of eukaryotic mRNA is the most common post-transcriptional modification to regulate mRNA maturation, stability, nuclear export, and translation efficiency [
14–
16]. It influences biological processes via m
6A methyltransferases (writers), m
6A demethylases (erasers), and m
6A-specific binding proteins (readers) [
17,
18]. By screening for RNA binding proteins (RBPs) that preferentially bind to m
6A-quantitative trait loci (QTL) regions, researchers have identified the TDP-43 encoding gene
TARDBP as a candidate m
6A reader gene [
19]. This finding was supported by recent findings indicating that TDP-43 may act as an indirect m
6A reader [
20]. Mechanistically, the aggregation of TDP-43 may possibly bind to the m
6A-modified mRNA via interacting with RBPs HNRNPA2B1 or YTHDF2, further leading to neurotoxic type I interferon responses [
20–
22]. Given that the mRNA transcripts encoding type I interferons are heavily m
6A-modified [
23], and that several genes encoding m
6A binding proteins are ALS-implicated risk genes, impaired mRNA metabolism and immune dysregulation are likely to converge on ALS pathogenesis.
Previous research on ALS has focused on non-cell autonomous neuronal loss caused by aberrant glial cell activation, whereas the roles played by infiltrated peripheral myeloid cells following blood–brain barrier impairment have not been fully explored [
24–
27]. Motor neurons are constantly exposed to cytokines produced by circulating immune cells via their distal motor axon and neuromuscular junction. In fact, the incidence of ALS with prior diagnosis of autoimmune disorders is significantly higher than expected, indicating a potential link between dysfunction in peripheral immune system and the development of ALS [
28]. This fact is consistent with the observation that the onset and progression of ALS are associated with innate and adaptive immunity, likely reflecting the unique pro-inflammatory gene expression profiles adopted by peripheral monocytes [
29], macrophages [
30], and T cells [
31]. Moreover, m
6A QTLs appear to be enriched for variants of complex traits related to blood cell and autoimmunity [
32]. Thus, clarifying the regulatory mechanism of dynamic mRNA methylation in peripheral immune cells could be a promising approach to elucidate ALS pathophysiology.
This study aims to examine the role of m6A methylation in peripheral immune cells in the pathogenesis of ALS through the implementation of an integrated approach that combines epi-transcriptomic and proteomic analyses. Specifically, the study aims to identify genes with differential methylation and expression patterns in peripheral immune cells and investigate their expression profiles in CNS by utilizing single-cell sequencing data derived from the primary motor cortex of patients with ALS and healthy controls. CellChat was employed to explore the altered intercellular communication network in ALS (Fig.1). The investigation revealed distinctive patterns of m6A modification in the peripheral immune cells of patients with ALS, which primarily affect cell migration. The results also suggested altered intercellular signaling between neurons and glial cells in ALS. These preliminary discoveries provide insights into the potential involvement of immune cell dysfunction in the pathogenesis of ALS and advocate for further functional validation in the direction of m6A-associated regulatory mechanisms. Overall, the integrated approach employed in this study represents a valuable basis for exploring the potential pathological significance of m6A methylation in ALS, shedding light on the aspects of this devastating disease that were previously underestimated.
2 Materials and methods
2.1 Subjects and sample collection
The study design and sample collection were approved by the Research Ethical Boards of Peking Union Medical College Hospital (PUMCH, No. JS-2624). Prior to participation, all subjects were informed, and they signed a written consent form or had a relative sign on their behalf. Blood samples were obtained from 16 patients diagnosed with clinically definite, probable, or probable laboratory-supported ALS in accordance with El Escorial criteria and from six matched healthy controls (Table S1). Only patients without signs of cognitive impairment were recruited to limit the effects of phenotypic heterogeneity, and the patients were stratified into two subgroups on the basis of the presence of bulbar symptoms at the time of sample collection (ALS_b for bulbar (n = 7) and ALS_s for spinal (n = 9)). Peripheral blood (10 mL) was collected using PAXgene® Blood RNA Tubes. Serum was separated through centrifugation at 3500 rpm for 10 min at 4 °C and then divided into aliquots. All samples were stored at −80 °C within 1 h of collection until use. The amount and purity of total RNA were measured with a NanoDrop ND-1000 spectrophotometer, and RNA integrity was assessed using denaturing agarose gel electrophoresis and Agilent Bioanalyzer 2100.
2.2 Immortalized cell lines and cell migration scratch assay
RAW264.7 murine macrophage cell line was obtained from ATCC and cultured in complete high-glucose Dulbecco’s Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin solution. The cells were maintained in a humidified atmosphere with 5% CO
2 at 37 °C and passaged every 2–3 days. For CX3CR1 stimulation and inhibition, RAW264.7 cells were seeded overnight onto six-well plates at a density of 5 × 10
5 cells/well and then treated with 1 μg/mL lipopolysaccharide (LPS, Solarbio, #L8880) for 4 h. Afterwards, the cells were incubated for 24 h in the presence of 0.2 μg/mL CX3CR1 ligand fractalkine (FKN, OriCell, #HECXP-01031) and 10 μM high-affinity selective CX3CR1 inhibitor AZD8797 (Chemegen, #C105220) [
33]. For cell migration scratch assay, RAW264.7 macrophages were seeded in six-well plates and grown to confluence. Scratches were made with a sterile 20 μL pipette tip after 4 h stimulation with LPS. The stimulated cells were rinsed two times with PBS and then subjected to serum-free medium with/without AZD8797. Images were taken at 0 and 12 h following the scratch, and the scratch areas were measured using ImageJ software [
34].
2.3 MeRIP-seq and MeRIP-qPCR
Peripheral blood cells were used to extract the total RNA, with 2.5 μg of the RNA fragmented into 100-nucleotide fragments through 5 min incubation at 94 °C by using NEBNext Magnesium RNA Fragmentation Module (NEB, #E6150S). Then, 10% of the fragmented RNA was set aside as input control, whereas the remainder was incubated with 0.2 μg anti-m6A polyclonal rabbit antibody (Synaptic Systems, #202003) that was conjugated to pre-washed Dynabeads Protein A (Thermo Fisher Scientific, #10002D) for 2 h at 4 °C with gentle rotation. After the incubation, the beads were washed three times with 1× RIP buffer (150 mM NaCl, 10 mM Tris-HCl (pH 7.5), and 0.05% Triton X-100), and the bound RNA was eluted from the beads and purified with TRIzol reagents (Thermo Fisher Scientific, #15596018) in accordance with the manufacturer’s instructions. Sequencing libraries were prepared from the input and immunoprecipitated RNA by using the SMARTer Stranded Total RNA-Seq Kit (Takara, #634413) and then sequenced on an Illumina NovaSeq 6000 platform in paired-end 150-base mode. After adapter sequences at the 3′ ends and low-quality bases (Q < 15) were removed with fastp (version 0.19.5), the remaining reads were evaluated with FastQC (version 0.11.4) and aligned to the human reference genome (GRCh38) by using STAR (version 2.7.7a) and HISAT2 (version 2.2.1). Peaks were identified using exomePeak2 (version 1.0.0) and visualized using Integrative Genomics Viewer (IGV). Homer (version 4.11) was used to perform motif enrichment with a motif length of six nucleotides. R package DESeq2 (version 1.10.1) was used to analyze differential peaks between groups. The calculated significance threshold (P≤ 0.05) of Fisher’s test was used to select significantly enriched GO terms on the basis of biological process and KEGG pathways by using clusterProfiler (version 2.4.2).
MeRIP-qPCR analysis was conducted on the basis of the predicted m6A sites by using SRAMP, and the input and immunoprecipitated RNA were prepared using the same protocol as described above but with scaled-down reagents. RT-qPCR analysis was then performed in accordance with the experimental procedure described below to validate the modification of m6A in CX3CR1.
2.4 RT-qPCR
A qScript cDNA Synthesis Kit (Quantabio, #95048-025) was used to prepare the cDNA, followed by RT-qPCR on a QuantStudio 12K Flex Real-Time PCR System (Life Technologies) using the SYBR Premix Ex Taq (Takara, # 4472903). The results were normalized to GAPDH, and the relative expression of mRNAs was quantified using the 2–∆∆Ct method. Details of the primers used are shown in Table S2.
2.5 RNA-seq
The total RNA from LPS-stimulated RAW264.7 cells, with or without FKN and AZD8797 priming, was extracted using TRI Reagent and fragmented. The BGISEQ platform was utilized to generate sequencing libraries, and the libraries with 150 bp paired-end reads (PE150) were sequenced to achieve a depth of around 45 million reads per sample. After the reads were filtered by removing adapter sequences and low-quality reads, the remaining reads were aligned to the “Mus musculus” reference genome (GCF_000001635.27_GRCm39) by using HISAT (version 2.0.4). RSEM (version 1.2.8) was used for all samples to quantify gene expression. DEseq2 was used for differential expression analysis. Pathway enrichment analysis of genes with significantly differential expression was conducted using Gene Set Enrichment Analysis (GSEA) [
35].
2.6 Serum FKN quantification (ELISA)
Blood samples were collected in Vacutainer Serum Tubes (BD Biosciences) from patients with ALS and healthy controls (n = 6). They were allowed to clot at room temperature before being separated by centrifugation (1000 g at room temperature for 10 min) within 4 h of collection and then stored at –80 °C until batch analysis. Soluble FKN levels were measured via ELISA (Human CX3CL1 ELISA KIT, #SEKH-0067, Solarbio) in accordance with the manufacturer’s instructions.
2.7 Liquid chromatography–mass spectrometry (LC–MS)
Serum samples were processed for protein analysis in the following manner. High-abundance proteins were depleted using High-Select Top 14 Abundant Protein Depletion Resin (Thermo Fisher, #A36369). The protein concentration was determined using the Bradford method. The protein extracts were then subjected to reduction with 25 mmol/L dithiothreitol at 37 °C for 60 min, followed by alkylation with 50 mmol/L iodoacetamide at room temperature for 30 min in the dark. The mixture was transferred to a 10 kDa ultrafiltration filter and washed with 20 and 50 mmol/L triethylammonium bicarbonate buffer. Trypsin digestion was performed overnight at 37 °C at a trypsin-to-protein mass ratio of 1:50. The resulting tryptic peptides were dried, resuspended in mobile phase A (2% (v/v) acetonitrile, 98% (v/v) ddH2O, pH 10), and centrifuged. The peptides were loaded onto a XBridge peptide BEH C18 column (4.6 × 15 mm, 3.5 μm, 130 Å) and eluted using stepwise injections of mobile phase B (98% acetonitrile, 2% ddH2O, pH 10) in the RIGOL L-3000 system (RIGOL, China). The chromatographic run lasted for 46 min with a flow rate of 1 mL/min, starting with 5% mobile phase B for 5 min, followed by a gradient from 5% to 30% mobile phase B over 31 min; a gradient from 30% to 95% mobile phase B over 5 min; and finally, a 5 min equilibration with 5% mobile phase B. Fractions were collected using the step gradients of mobile phase B at 1 min intervals.
For MS analysis, 1 μg of the collected samples was loaded onto an EASY-nLC1000 system connected to a Q Exactive HF mass spectrometer (Thermo Scientific). Peptides were eluted using a binary solvent system consisting of 99.9% H2O, 0.1% formic acid (phase A), 99.9% ACN, and 0.1% formic acid (phase B) at a flow rate of 0.6 μL/min. The elution gradient comprised 6%–30% phase B over 69 min, followed by 30%–42% phase B over 12 min, 42%–100% phase B over 4 min, and a 5 min wash at 100% phase B. The eluent was introduced to an Orbitrap Exploris 240 mass spectrometer via an EASY-Spray ion source with specific ionization parameters (spray voltage, 2.2 kV; capillary temperature, 320 °C; declustering potential, 100 V). For data-dependent acquisition (DDA) MS, a full-scan MS was conducted with specific settings (scan range, 300–1500 m/z; isolation window, 2 m/z; max injection time, 20 ms; AGC target, 3 × 106; collision energy, 30%; resolution, 60 k), followed by 20 MS/MS scans (max injection time, 22 ms; dynamic exclusion duration, 45 s; AGC target, 7.5 × 104; included charge states, 2–6; resolution, 15 k). The DDA spectra were characterized using Proteome Discoverer (version 2.3.0.523) analysis software with default settings. The search criteria included carbamidomethylation (cysteine) as a fixed modification, and oxidation (methionine) and acetyl (protein N terminus) as variable modifications. The precursor mass tolerance was 10 ppm, and the fragment-ion mass tolerance was 0.02 Da. For data-independent acquisition (DIA) MS, an MS1 scan was conducted with specific settings (scan range, 300–1300 m/z; AGC target, 3 × 106; collision energy, 32%; resolution, 30 k), followed by a MS2 scan with multiple scan windows. The DIA data were analyzed using a Spectronaut pulsar (Biognosys, version 15.0.210615.50606) with default settings, including retention time prediction and correction, mass calibration, decoy generation, interference correction, and FDR estimation. The raw files were converted into Spectronaut file format to analyze the DIA runs with the spectral library, calibrated in the retention time dimension, and then used for targeted data analysis with the spectral library without additional recalibration.
2.8 Parallel reaction monitoring (PRM)
In the independent cohort of eight patients with ALS and eight healthy controls (Table S3), selected proteins from the discovery cohort were selected. Peptide samples were separated using a reverse-phase column (Dr. Maisch GmbH) with a 250 mL/min flow rate of increasing buffer B concentration (2%–60%) over 98 min on an HPLC system (Thermo Fisher Scientific). The separated peptides were analyzed on a Q-Exactive HFx (Thermo Fisher Scientific) using PRM settings of 30 000 resolutions, 2 × 105 AGC target, 1.6 m/z isolation window, and 60 ms maximum ion injection time. Data analysis was conducted using Skyline software. Peaks were selected manually on the basis of retention time and dot product, and the total intensity value of the correct peak area was extracted for peptide quantification.
2.9 Analysis of single-cell mRNA sequencing data
The gene-barcode matrix of the primary motor cortex’s single-cell sequencing data derived from 17 patients with ALS and 17 pathologically normal (PN) controls (GSE174332) [
36] was analyzed using Seurat R toolkit (version 4.2.1) [
37]. The quality control parameters included < 50 genes/cell, < 3 cells per genes, and > 7% mitochondrial genes. PCA was performed on the top 2000 most variable genes, and the first 15 principal components were used for k-means clustering and k-nearest neighbor graph construction with a clustering resolution parameter of 0.5. Cell clusters were visualized using the Uniform Approximation and Projection (UMAP) method and annotated on the basis of cell type-specific markers. Differential gene testing was conducted using the FindMarkers function with non-parametric Wilcoxon rank sum test. GO and KEGG enrichment analysis was performed to summarize the differential genes functionally. The DM_score was calculated using the AddModuleScore function in Seurat R package. Random Forest regression and LASSO regression were used to analyze the data by using the randomForestSRC and glmnet R packages, respectively. CellChat (version 1.6.1) was used to infer intercellular signaling networks on the basis of the curated ligand–receptor interaction database [
38].
2.10 Statistical analysis
Data were statistically analyzed using GraphPad Prism (version 7.0) and R (version 4.2.2). Unpaired two-tailed Student’s t-test or two-way ANOVA was used to compare groups as appropriate. The results were presented as means ± standard deviation (SD) with at least three biological replicates. A P value of less than 0.05 was considered statistically significant.
3 Results
3.1 Widespread mRNA hypermethylation in peripheral immune cells from patients with ALS
Transcriptome-wide m6A modification profiling of peripheral blood cells from patients with ALS and healthy controls was conducted to explore the potential role of m6A methylation in ALS pathogenesis. A total of 36.38–62.23 million reads were generated from each m6A-seq library after quality control. The results revealed that the distribution pattern of m6A modification across genes was similar between the two groups, with most m6A peaks preferentially located in the transcription start site and stop codon site (Fig.2), and most methylated mRNAs containing one or two peaks (Fig.2). The peak motif observed in this study was consistent with the known consensus m6A methylation motif (Fig.2). Moreover, the ALS samples had a greater number of hypermethylated peaks than the controls, whereas fewer hypomethylated peaks were identified (log2|FC| > log2(1.5), adjusted P < 0.05; Fig.2). The candidate genes that were potentially modulated by RNA methylation were assessed through the integration of m6A methylation and RNA expression profiles (Fig.2). Specifically, 109 and 160 deferentially expressed genes with differential m6A methylation peaks were identified in the ALS_b and ALS_s samples, respectively (Fig.2 and Table S4). These results demonstrated the probable involvement of m6A methylation in the regulation of gene expression, thus highlighting the significance of investigating RNA modification in disease pathogenesis.
3.2 Candidate m6A-regulated genes screened by differential methylation and expression
GO and KEGG enrichment analyses were conducted to further explore the potential biological implications of differential methylation in the context of ALS. The results unveiled that the differentially methylated genes are associated with various biological processes, including chromatin modification, histone modification, and endocytosis and inflammatory signaling pathways (Fig.3 and 3B). The functional pathway analyses did not reveal any significant differences between the two ALS subgroups, thus allowing this study to focus on common differentially methylated and expressed genes that may be relevant to ALS pathogenesis. A total of 15 genes that were differentially methylated and expressed in both ALS subgroups were identified, some of which are crucial regulators of immune cells and neurons (Fig.3). Next, qRT–PCR to verify the expression levels of several genes selected on the basis of their biological relevance (Fig.3). This study focused on
CX3CR1 given the evidence supporting its involvement in ALS pathogenesis. The examination of the read density by using IGV showed enhanced m
6A methylation on the
CX3CR1 transcript in ALS samples (Fig.3). Consistent with this finding, the SRAMP browser tool [
39] predicted several m
6A modification sites located on the
CX3CR1 RNA sequence, with high confidence scores, which overlapped with the sequencing results. The MeRIP–qPCR analysis further confirmed the differential methylation of
CX3CR1 at the selected region (
P = 0.0037, Fig.3). Meanwhile, the online m
6A target database suggested that
Mettl3 knockout and
Mettl14 knockdown are associated with increased
CX3CR1 gene expression in different cell lines [
40]. The serum levels of the CX3CR1 ligand FKN were assessed, and no statistically significant distinctions between patients with ALS and controls was found (Fig.3). These findings underscored the probable involvement of m
6A-mediated regulation of
CX3CR1 in the pathogenesis of ALS.
3.3 CX3CR1 signaling is involved in regulating macrophage migration
Due to the prevalence of immune dysregulation characterized by increased proinflammatory macrophages in patients with ALS [
29] and the active involvement of m
6A modification in regulating macrophage functions [
41], further investigations into how the macrophage cell line RAW264.7 respond to stimulation and inhibition of CX3CR1 signaling
in vitro were conducted. LPS-primed RAW264.7 cells were treated with either FKN or AZD8797 for 24 h before subjecting them to mRNA-seq to detect transcriptomic changes (log
2|FC| > log
2(1.5), adjusted
P < 0.05) for pathway enrichment analyses. Consistent with the known biological functions of CX3CR1, the KEGG results indicated that activating and blocking CX3CR1 mainly affected cytokine–cytokine receptor interaction (Fig.4). In line with this finding, GSEA using GO gene sets suggested that CX3CR1 signaling was associated with cell mobility, chemotaxis, and cellular response to interferons (Fig.4 and 4C). The differential expression of several immune-associated genes selected based on the sequencing results and literature were verified by RT-qPCR (Fig.4). The scratch assays showed that cell migration was reduced by AZD8797 treatment (Fig.4). Collectively, these findings suggested that CX3CR1 signaling is likely involved in the pathogenesis of ALS by regulating the migration function of peripheral immune cells.
3.4 Serum proteomic analyses supported the altered cell migration function in ALS
Previous studies suggested that proteome homeostasis disturbance is a key factor underlying cellular dysfunction in ALS pathogenesis [
42]. LC–MS was employed to analyze the serum proteome of patients with ALS to investigate the association between altered immune cell phenotypes and differential m
6A modification at the protein level. A total of 3122 distinct peptides (detectable in at least 50% of all samples) representing 711 proteins were identified via DIA. Specifically, 33 proteins were significantly altered in ALS_b (27 downregulated and six upregulated), and 39 proteins were significantly altered in ALS_s (32 downregulated and seven upregulated) compared with controls (log
2|FC| > log
2(1.3), adjusted
P value < 0.05; Table S5). The differential proteins accurately clustered the subjects, thereby validating the arbitrary thresholds imposed for filtering (Fig.5). Consistent with previous findings in ALS and frontotemporal dementia (FTD) serum samples, the present study identified downregulated APOA2, COMP, and CRTAC1 and upregulated S1008A8 [
43,
44]. By integrating the results with m
6A-seq data, several identified proteins were observed to be differentially m
6A-modified, thus supporting the potential regulatory functions of m
6A modification at the protein level (Fig.5). The STRING web tool was used to investigate the possible protein–protein interactions (PPIs) of the differential proteins (Fig.5). The analysis revealed that the differentially expressed proteins were enriched for extracellular matrix organization, cell adhesion, and regulation of collagen biosynthetic process. A notable detail that several network nodes were differentially m
6A-modified, suggesting a potential involvement of m
6A modification in regulating cell motility, although such observation alone did not establish a direct causal link. The LC–MS results were validated in an independent cohort via PRM. This assay enabled the determination of protein levels for 23 specific proteins in serum samples, with selection guided by the results of PPI pathway analysis. Among these proteins, statistically significant differences were observed in the serum levels of ENG, CDH13, TNXB, CLEC3B, and DEFA1 (Fig.5), consistent with the MS results.
3.5 Expression patterns of differentially methylated genes in ALS motor cortex
The observation of widespread RNA hypermethylation in post-mortem spinal cords of patients with ALS led to the further investigation of whether the differentially methylated genes identified in peripheral immune cells may affect cellular functions within CNS. ScRNA-seq data derived from the human primary motor cortex of patients with ALS and controls were analyzed [
37]. Neurons (excitatory and inhibitory), glial cells, and vascular cells were identified on the basis of canonical CNS cell markers before further sub-clustering (Fig.6). The average expression levels of the differentially methylated genes identified in peripheral immune cells (DM_score) were calculated using Seurat’s AddModuleScore function. The DM_score was highly heterogenous in glial cell populations (Fig.6), with microglia displaying the highest degree of heterogeneity (Fig.6). The DM_score was significantly lower in ALS-derived cells, particularly for microglia (Fig.6), indicating that on average, these genes were differentially expressed in ALS primary motor cortex. Random Forest and LASSO regression analyses were further conducted to refine the candidate genes most likely involved in ALS pathogenesis (Fig.6 and 6F). Eleven overlapping genes were identified, several of which were involved in regulating cell mobility and immune cell infiltration (
CORO1B [
45],
ARHGEF18 [
46], and
ISYNA1 [
47]), as shown in Fig.6. Notably, these genes were top-ranked in the Random Forest regression model, supporting the critical role of cell–cell interaction in ALS pathogenesis. The findings suggested that differential m
6A modification may similarly regulate immune cell characteristics in CNS and the periphery, and intercellular communication in CNS is an important aspect requiring further clarification.
3.6 Altered intercellular signaling network between neuronal and non-neuronal cells in ALS
Given the potential involvement of m6A-mediated regulation in influencing the migration of peripheral immune cells in the context of ALS pathogenesis, the perturbed patterns of intercellular communication existing between neuronal and immune cell populations within CNS were explored. The CellChat algorithm was utilized to conduct a comparative analysis of signaling patterns in the motor cortex of ALS and control subjects. The global communication between neuronal and non-neuronal cell clusters was quantified. The inferred number of interactions in ALS generally increased, whereas the interaction strength in inhibitory neurons with oligodendrocytes and excitatory neurons decreased (Fig.7). By calculating the information flow for each signaling pathway, which is defined as the sum of communication probability among all pairs of cell groups in the inferred network, prominent changes in the pathway information flow in ALS were observed as follows: turned off (e.g., ENHO, KIT, and CCK), decreased (e.g., TGFb, GAS, and CX3C), or increased (e.g., ANGPT, EDN, and VEGF; Fig.7). Then, the Euclidean distance was computed to measure dissimilarity between pairs of signaling pathways shared by ALS and PN. Significant differences were found in the communication network architecture of signaling pathways for ANGPT, PROS, VEGF, CX3C, and TGFb in ALS compared with PN (Fig.7), implying that these pathways are greatly altered in ALS (larger Euclidean distance). Heatmaps of the pathway-specific signaling flows further indicated that the inferred alterations mainly involved excitatory and inhibitory neurons (Fig.7). Excitatory neuron subtypes were the dominant sources of CX3C ligand to microglia in ALS and PN, but a minor contribution of signals from inhibitory basket cells was found in PN, which was absent from ALS cells (Fig.7 and 7F). Similarly, decreased GAS6-AXL signaling was observed between microglia and inhibitory neurons in ALS (Fig.7 and 7H). A pattern recognition method based on nonnegative matrix factorization was applied to further explore how these implicated signaling pathways and cell groups coordinate to function. The resulting Sankey plots revealed four patterns for incoming signaling (Fig.7) and four patterns for outgoing signaling (Fig.7). The communication patterns of target cells showed that the incoming microglia and oligodendrocyte signals were dominated by pattern #3, which was characterized by multiple signaling pathways regulating CNS inflammatory responses, such as CX3C, PSAP, PROS, and VISFATIN, that were found to be reduced in ALS. The outgoing communication pattern of these pathways was featured by pattern #1, which was contributed almost exclusively by excitatory neurons. In summary, while most incoming microglia signaling was contributed by excitatory neurons, reduced levels of signaling, such as CX3C from inhibitory neurons, may play critical roles in regulating microglia phenotype during disease pathogenesis. These results highlighted the altered intercellular communication between neuronal and non-neuronal cells in ALS, although direct evaluation of m6A methylation in CNS is warranted to confirm its involvement in the decreased intercellular signaling between neurons and glial cells.
4 Discussion
RNA destabilization and dysregulated induction of immune cells are recurrent themes in the pathogenesis of ALS [
48,
49]. RNA m
6A modification has notably emerged as a prominent player in the regulation of innate and adaptive immune responses, particularly in an interferon signaling-dependent manner [
50]. The MeRIP-seq profiling of peripheral immune cells in the present study revealed that most differentially m
6A-modified genes were hypermethylated in ALS, aligning with the recent findings of widespread m
6A RNA hypermethylation in post-mortem spinal cord samples from patients with ALS [
20]. A notable detail that conflicting results were reported in iPSC-differentiated neurons derived from patients with
C9orf72-ALS/FTD. In this context, global m
6A hypomethylation has been observed to lead to an increased accumulation of
C9orf72 repeat expansions and poly-dipeptides [
51]. These findings suggest that dysregulated m
6A modification, which actively participates in ALS pathogenesis, may exert its effect on mRNA metabolism in a context-dependent manner. For example, m
6A-mediated RNA regulation exhibits distinct outcomes, with the binding of m
6A reader YTHDF2 promoting mRNA degradation [
52], whereas IGF2BPs stabilize methylated transcripts [
53]. Therefore, m
6A modification may serve as an upstream regulator of multiple immune pathways in ALS, thus highlighting its potential role in the modulation of immune responses associated with the disease. However, determining the predominant causal role among these pathways, either individually or in combination, remains a challenge.
The integrated transcriptomic analyses of peripheral blood samples provided some preliminary insights. Among the genes that were differentially methylated and expressed in both ALS subgroups, several were implicated in pathways relevant to ALS pathophysiology. For example, m
6A methylation is actively involved in the homeostasis of peripheral immune cells by modulating the expression of key regulators on JAK–STAT signaling pathways, such as SOCS family members [
54]. Overexpression of
SOCS genes attenuates IL-7- dependent CD4
+ T cell proliferation and induces defective regulatory T cell (Treg) activity [
55]. Myeloid lineage-restricted deletion of
METTL3 results in a significant increase in
DDIT4 mRNA levels, which is associated with immunity-driven nonalcoholic fatty liver disease [
56,
57]. Overexpression of
TSC22D3 in mature Tregs can impair their immunoregulatory function during intestinal inflammation [
58], and
CYP4F3 is involved in inflammatory processes by specifically metabolizing the neutrophil chemoattractant
LTB4 [
59]. In CNS,
DAAM2 plays an essential role in glial cell morphogenesis and myelination by regulating the Wnt signaling pathway [
60–
62], and
CDK5R1 (also known as p35) is associated with neuron tauopathies and axon outgrowth impairment [
63].
Previous studies suggested that
CX3CR1 polymorphisms may serve as disease-modifying factors in ALS [
64,
65], and cells expressing
CX3CR1V249I/T280M exhibited defective signaling and increased adhesive capacity to FKN [
66]. As an exclusive receptor for FKN,
CX3CR1 is primarily expressed by microglia in CNS and certain peripheral immune cells such as perivascular macrophages and monocytes [
66,
67]. Its primary role in CNS is to suppress proinflammatory activities, as disruption of CX3CR1 signaling in systemic LPS-primed
SOD1G93A mouse models leads to aberrant microglial activation and neurotoxicity [
68]. In the periphery, CX3CR1 plays a critical role in cell adhesion, ensuring the retention of leukocytes within tissues [
69]. Reduced
CX3CR1 expression is likely to disrupt immune cell trapping and exacerbate neuronal loss in ALS. This finding aligns with the altered immune cell migration function observed in the network-based proteomic analysis of ALS serum samples. The experiments involving the activation and blockade of CX3CR1 in LPS-stimulated RAW264.7 cells in the present study resulted in transcriptomic changes associated with chemokine-mediated signaling pathways and cell mobility. There is a notable detail that LPS treatment was found to reduce
CX3CR1 expression [
70,
71] and enhance
METTL3 expression and overall m
6A levels in macrophages [
72], dendritic cells [
73], and dental pulp cells [
74]. Given that m
6A methylation regulates RNA stability, the observed hypermethylation of
CX3CR1 mRNA in ALS samples may indicate accelerated decay and decreased expression. Therefore, although further functional validation is necessary, m
6A modification may serve as a remarkable regulator of macrophage activation in response to LPS.
Apart from macrophages, infiltration of monocytes [
75] and NK cells [
76] into the motor cortex of TDP-43
A315T mice and patients with TDP-43 pathology has been noted, although the extent to which the peripheral immune cell phenotypes may influence ALS neuroinflammation remains inconclusive. In this study, the methylation patterns of CNS immune cells were not directly quantified, so the average expression of differentially methylated genes was correlated to the scRNA-seq data set of primary motor cortex. The results indicated that these m
6A-modified genes were differentially expressed in ALS samples, with microglia demonstrating the most prominent differences. CellChat further suggested that the alteration of intricate cellular communication between neuronal and non-neuronal cells may be a crucial aspect of ALS pathogenesis. These results confirmed that CX3C signaling was reduced between microglia and inhibitory neurons in ALS motor cortex. In addition to CX3C signaling, disturbance of other signaling pathways sharing similar communication pattern may induce neurodegeneration, possibly via immune-associated pathways. For example, reduced GAS6-AXL signaling was observed between microglia and inhibitory neurons in ALS, which is a critical regulator of microglial inflammatory responses in Parkinson’s disease [
77] and Alzheimer’s disease [
78] mouse models. Similarly, reduced PSAP signaling was observed in ALS, the diminished trafficking of which underlies FTD pathogenesis [
79]. Therefore, the differential methylation detected in the peripheral immune cells may serve as a reflection of the CNS transcriptome, the alteration of which could likely induce dysregulated intercellular communication in the motor cortex (Fig.8).
An important limitation of this study is the lack of direct evidence demonstrating the biological consequences of differential m
6A methylation in CNS tissue. Cellular models combined with m
6A methyltransferase gene knockout assays using induced pluripotent stem cell (iPSC) from patients with ALS may provide useful insights, because neuron–glial cell crosstalk is essential for inducing neurotoxicity in ALS [
80]. However,
in-vitro cells display significant transcriptional changes compared with
in-vivo cells, thus making direct evidence derived from animal models necessary to firmly establish any causal links, although trans-species differences could be another issue that needs to be addressed. Nevertheless, the bioinformatic analyses of the human motor cortex provides encouraging results for carrying out further functional validation. Given the relatively small sample size of this study, sequencing data from a larger cohort for validation in the future are desirable.
In conclusion, although significant progress has been made in understanding the role of m6A modification in immune phenotypes, further efforts are needed to understand how aberrant m6A-mediated gene regulation contributes to the etiology of complex diseases like ALS. In this study, the m6A methylation pattern of peripheral immune cells in ALS was profiled, and the differential methylation was correlated with serum proteome and CNS transcriptome. The integrated analyses highlighted the previously underexplored m6A-mediated RNA regulation in ALS pathogenesis, which may have important therapeutic implications in the future.