Research Paper Volume 14, Issue 16 pp 6554—6566

Distinct microglia alternative splicing in Alzheimer's disease

Yanjun Lu1, *, , Lu Tan2, *, , Jiazhao Xie3, , Liming Cheng1, , Xiong Wang1, ,

  • 1 Department of Laboratory Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
  • 2 Key Laboratory for Molecular Diagnosis of Hubei Province, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430014, China
  • 3 Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
* Equal contribution

Received: May 11, 2022       Accepted: August 3, 2022       Published: August 23, 2022      

https://doi.org/10.18632/aging.204223
How to Cite

Copyright: © 2022 Lu et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Numerous alternative splicing (AS) events have been documented in Alzheimer's disease (AD). However, cell type-specific AS analysis is still lacking. We described AS events in the hippocampal microglia sorted by CD45 and CD11b from Aβ precursor protein (APP) and non-transgenic (Ntg) mice. GSE171195 dataset was downloaded from GEO database, aligned to GRCm39 genome. Skipped exon (SE), alternative 3’SS (A3SS), retained intron (RI), alternative 5’SS (A5SS), and mutually exclusive exons (MXE) were evaluated using rMATS and maser. Differential expressed genes or transcripts were analyzed via limma. Gene ontology and correlation analyses were performed with clusterProfiler and ggcorrplot R packages. 36,340 raw counts of AS were identified, and 95 significant AS events were eventually selected with strict criteria: (1) average coverage >5; (2) delta percent spliced in >0.1. SE was the most common AS events (68.42%), followed by A3SS and RI. Autophagy genes were mainly spliced in SE events, actin depolymerization genes spliced in A3SS events, while synaptic plasticity related genes were mainly spliced in RI pattern. These significant AS events may be regulated by dysregulated splicing factors in AD. In conclusion, we revealed microglia specific AS events in AD, and our study provides novel pathological mechanisms in the pathogenesis of AD.

Introduction

Splicing converts precursor mRNA into mature mRNA via spliceosome, a multiprotein-RNA complex with highly specific and stepwise interactions. The spliceosome contains 5 small nuclear ribonucleoproteins (snRNPs; U1 to U6 snRNPs). U1 snRNP binds to 5′ splice site, and U2 snRNP binds to 3′ splice site and polypyrimidine sequences. Next, U5, U4/U6 were recruited to complete intron excision and exon linkage mainly following the “GU-AG” rule [1]. Alternative splicing (AS) enables cells to diversify their proteome by generating distinct characteristics to different protein isoforms through different combination of exons in the mRNA, leading to complex biological functions under conditions of external and internal stimuli. AS plays essential roles in cell differentiation and organ development t [2]. Generally, five basic modes of AS are classified: alternative 5′ splice site (A5SS), skipped exon (SE), mutually exclusive exons (MXE), retained intron (RI), and alternative 3′ splice site (A3SS) [3]. AS dynamically changes during aging, and aberrant AS events have been observed in numerous diseases including cancer through providing malignant protein isoforms [4]. Carcinogenesis involves cellular proliferation, invasion and metastasis, induction of angiogenesis, and immune escape. Recurrent somatic mutations of the splicing machinery components have been observed in human solid tumors and hematological malignancies. AS participates in these processes by regulating the alternative expression of numerous oncogenic or tumor suppressor genes [5].

Aberrant AS events have been characterized in Alzheimer’s disease (AD), the most frequent cause of dementia. AD is hallmarked by β-amyloid (Aβ) plaques and hyperphosphorylated tau tangles. Both genetic and environmental risk factors contribute to AD pathogenesis [6]. The triggering receptor expressed in myeloid cells 2 (TREM2) mediates inflammatory responses, microglia activation, and phagocytosis of apoptotic neurons. TREM2 is involved in tau accumulation and Aβ plaque formation and therefore strongly affects risk of AD [7]. TREM2 rare variants caused exon 2 skipping, and activated immune-related functional pathways in AD [8]. Nuclear speckle specific hnRNP D-like prevented AD-related cognitive decline via modulating AS of synaptic gene calmodulin kinase-like vesicle-associated (CAMKV) [9]. Numerous AS events mainly including SE and RI have been documented in both AD human and animal models [10, 11].

These studies mainly focused on different models or brain regions. We systematically described AS events in the hippocampal microglia (CD45int CD11b+) sorted with FACSAria II (BD) of Aβ precursor protein (APP) and non-transgenic (Ntg) mice. We revealed hippocampal microglia cell type-specific AS events in AD.

Results

Global summary of alternative splicing events

A total of 36,340 AS events were initially identified in GSE171195 dataset (Figure 1A). Eventually, 95 significant AS events were collected (Figure 1B, Table 1). The principal component analysis (PCA) revealed that these AS events clearly classified APP from Ntg group (Figure 1C). Increased PSI was found in A3SS and SE in APP group, while decreased PSI was found in A3SS and global events (Figure 1D, 1E). Top two events in each AS event were included in Table 2, and all 95 significant AS events were shown in Supplementary Table 1.

Global summary of AS events. (A) Raw counts in each AS event. (B) Number and ratio of each significant AS event. (C) The PCA plot based on PSI values. APP and Ntg groups were clearly classified. (D) The percent of ΔPSI in each and global AS events. (E) Distribution of ΔPSI in each type AS events.

Figure 1. Global summary of AS events. (A) Raw counts in each AS event. (B) Number and ratio of each significant AS event. (C) The PCA plot based on PSI values. APP and Ntg groups were clearly classified. (D) The percent of ΔPSI in each and global AS events. (E) Distribution of ΔPSI in each type AS events.

Table 1. Event summary.

Event typeTotal events junction countsAverage coverage > 5FDR < 0.05, deltaPSI > 0.1Ratio in NtgRatio in APP
A3SS343324131111.60%11.50%
A5SS2096133779.30%5.80%
SE24944198666565.10%71.20%
RI295620341011.60%9.60%
MXE2911246522.30%1.90%
Total363402811595100.00%100.00%

Table 2. Top two events in each type of AS pattern.

IDP valueFDRΔPSIGeneType
39911.39E-070.0002380.15Tamm41A3SS
37562.60E-070.000297−0.167Ube2fA3SS
332.02E-070.000211−0.145IncenpA5SS
20202.34E-060.0016370.101Dcun1d2A5SS
4824.45E-091.23E-050.298RnlsSE
297531.63E-083.12E-050.304Rcor3SE
16052.91E-060.000782−0.127Rbm3RI
39268.27E-050.009775−0.101Myl6RI
32227.50E-112.18E-07−0.106Pld3MXE
33016.08E-050.022130.25ErbinMXE

Analysis of SE events

SE events ranked the most frequent AS events, accounting for 68.42% (Figure 1B). GO analysis of genes involved in significant SE events were enriched in regulation of autophagy (Figure 2A, 2B), including eukaryotic translation initiation factor 2 alpha kinase 4 (Eif2ak4) and two pore segment channel 2 (Tpcn2). Eif2ak4 tended to harbor longer exons, while Tpcn2 tended to harbor shorter exons in APP (Figure 2C, 2D).

Analysis of SE events. (A) Significant GO terms enriched in genes involved in SE events. (B) Cnetplot revealed genes in these enriched GO terms. Eif2ak4 colored in red indicated increased PSI level, while Tpcn2 colored in green represented decreased PSI level. (C, D) The detailed sashimi plots for Eif2ak4 and Tpcn2.

Figure 2. Analysis of SE events. (A) Significant GO terms enriched in genes involved in SE events. (B) Cnetplot revealed genes in these enriched GO terms. Eif2ak4 colored in red indicated increased PSI level, while Tpcn2 colored in green represented decreased PSI level. (C, D) The detailed sashimi plots for Eif2ak4 and Tpcn2.

Analysis of A3SS events

A total of 11 significant A3SS events were identified and ranked top 2 AS events (Figure 1B). GO enrichment revealed that genes with significant A3SS events were enriched in actin polymerization and depolymerization, including microtubule associated monooxygenase, calponin and LIM domain containing 1 (Mical1), and adducin 1 (Add1) (Figure 3A, 3B). The sashimi plot showed decreased PSI and a shorter transcript for Mical1 (Figure 3C), whereas increased PSI for Add1, indicating a longer transcript (Figure 3D).

Analysis of A3SS events. (A) Significant GO terms enriched in genes involved in A3SS events. (B) Cnetplot revealed genes in these enriched GO terms. Mical1 colored in red indicated increased PSI level, while Add1 colored in green represented decreased PSI level. (C, D) The detailed sashimi plots for Mical1 and Add1.

Figure 3. Analysis of A3SS events. (A) Significant GO terms enriched in genes involved in A3SS events. (B) Cnetplot revealed genes in these enriched GO terms. Mical1 colored in red indicated increased PSI level, while Add1 colored in green represented decreased PSI level. (C, D) The detailed sashimi plots for Mical1 and Add1.

Analysis of RI events

RI is third most common events in this study (Figure 1B). The PSI value referred to level of retained intron. GO analysis revealed enrichment of RNA splicing as well as synaptic plasticity including axonogenesis, neurotransmitter secretion, neuron projection and axon guidance (Figure 4A, 4B). Glypican 2 (Gpc2) showed increased RI level and enriched in neuron projection (Figure 4C). RNA binding motif protein 3 (Rbm3) showed decreased RI level and enriched in RNA splicing (Figure 4D).

Analysis of RI events. (A) Significant GO terms enriched in genes involved in RI events. (B) Cnetplot revealed genes in these enriched GO terms. Gpc2 colored in red indicated increased PSI level, while Rbm3 colored in green represented decreased PSI level. (C, D) The detailed sashimi plots for Gpc2 and Rbm3.

Figure 4. Analysis of RI events. (A) Significant GO terms enriched in genes involved in RI events. (B) Cnetplot revealed genes in these enriched GO terms. Gpc2 colored in red indicated increased PSI level, while Rbm3 colored in green represented decreased PSI level. (C, D) The detailed sashimi plots for Gpc2 and Rbm3.

Differentially expressed genes and transcript

Aberrantly spliced mRNAs would be degraded by nonsense-mediated mRNA decay (NMD) pathway. However, some mRNAs may bypass NMD and be translated. Differential expressed genes and transcripts were examined. These samples were well correlated within group (Figure 5A). 583 up- and 310 down-regulated genes were identified (Figure 5B, Supplementary Table 2). Only two genes with significant AS events showed significant difference between APP and Ntg groups on gene level (Figure 5C). On transcript level, 1225 up- and 756 down-regulated transcripts were identified (Supplementary Table 3). 17 out of the 26 transcripts, whose host genes with significant AS events, were involved in SE event, 6 transcripts were involved in RI event (Table 3). Several genes with significant AS events harbored transcripts with opposite changes, including Rbm3 and Azi2 (Figure 5D, Table 3). Given Azi2 for instance, SE event resulted in a longer transcript (ENSMUST00000130735) and a shorter transcript (ENSMUST00000133814) (Figure 5E). In SE event, Azi2 showed increased PSI for the longer transcript in APP group (Figure 5F). Consistently, the longer ENSMUST00000130735 with increased PSI in APP group was elevated, while the shorter ENSMUST00000133814 with decreased PSI in APP group showed suppressed expression (Figure 5G). However, the gene level expression of Azi2 showed no significant difference (Figure 5H).

Differentially genes and transcripts analysis. (A) Correlation analysis with gene expression matrix. (B) Heatmap of dysregulated genes between APP and Ntg group. (C) Volcano plot of dysregulated genes. The black circle indicated genes with significant AS event. (D) Volcano plot of dysregulated transcripts. Genes harbored different transcripts with opposite changes were labeled. (E) Genomic information of Azi2. Red arrow indicated the longer transcript with coding ability and SE event, and the green arrow revealed the shorter transcript with RI. (F) The detailed sashimi plots for Azi2. (G, H) Relative expression of Azi2 in gene and transcript levels. ENSMUST00000130735 represented longer Azi2 transcript, and ENSMUST00000133814 represented the shorter transcript.

Figure 5. Differentially genes and transcripts analysis. (A) Correlation analysis with gene expression matrix. (B) Heatmap of dysregulated genes between APP and Ntg group. (C) Volcano plot of dysregulated genes. The black circle indicated genes with significant AS event. (D) Volcano plot of dysregulated transcripts. Genes harbored different transcripts with opposite changes were labeled. (E) Genomic information of Azi2. Red arrow indicated the longer transcript with coding ability and SE event, and the green arrow revealed the shorter transcript with RI. (F) The detailed sashimi plots for Azi2. (G, H) Relative expression of Azi2 in gene and transcript levels. ENSMUST00000130735 represented longer Azi2 transcript, and ENSMUST00000133814 represented the shorter transcript.

Table 3. Dysregulated transcripts involved in significant AS genes.

IDGeneTranscriptlogFCP.ValueChangeAS type
3492Il4i1ENSMUST000000330151.4021531.43E-06UPSE
7070Lrrc56ENSMUST000000704580.7380230.024408UPSE
11659Rbm3ENSMUST000001156210.6473110.020246UPRI
11784Pld3ENSMUST000001170950.7394581.60E-07UPMXE
13865Eme2ENSMUST00000128953−0.918560.006028DOWNRI
14316Azi2ENSMUST000001307350.5855340.034119UPSE
14853Nfat5ENSMUST00000133026−0.683010.02389DOWNSE
15031Azi2ENSMUST00000133814−0.995080.039242DOWNSE
15298Rnf135ENSMUST000001349090.9234430.038781UPSE
15611Arhgef11ENSMUST00000136072−1.03610.038672DOWNSE
16477Il15raENSMUST000001397741.3517810.009183UPSE
16644St7lENSMUST00000140457−1.020250.041252DOWNSE
17009Rbm3ENSMUST00000141925−0.682970.0309DOWNRI
20175Pld3ENSMUST000001552871.2007792.63E-06UPMXE
20796Adck5ENSMUST00000159949−1.107560.029743DOWNRI
21734Tmem176bENSMUST000001647330.5941220.031092UPSE
22481Ankrd13dENSMUST00000170283−1.091350.03766DOWNSE
23261Pknox1ENSMUST000001758061.2527920.006284UPSE
246192410002F23RikENSMUST000001881110.9200560.002927UPSE
26213Krit1ENSMUST00000200386−0.590630.043742DOWNA3SS
26680Tmem176bENSMUST000002032290.5874060.049277UPSE
26995EmsyENSMUST00000205331−0.884220.012687DOWNSE
27595Tpcn2ENSMUST00000209047−1.101090.011942DOWNSE
27876Wdr74ENSMUST000002105920.9036490.02283UPRI
30538Camk2gENSMUST00000225958−0.657080.038834DOWNSE
32057Sema4gENSMUST00000235312−1.102670.009556DOWNRI

Analysis of upstream splicing factors

AS events could be regulated by a number of splicing factors (SFs). The expression of 280 SFs obtained from previous study was examined (Supplementary Table 4) [12]. A total of 37 transcripts involved 30 SF genes were significantly dysregulated between APP and Ntg groups (Figure 6A, Table 4). The PPI networks were constructed using String and Cytoscape (Figure 6B). The first and second clusters of these SFs were identified using Cytoscape with MCODE app (Figure 6C, 6D). The correlation of the two clusters and significantly dysregulated transcripts whose host genes were involved in significant AS events were performed (Figure 6E, 6F).

Analysis of upstream splicing factors. (A) volcano plot of dysregulated splicing factors. (B) PPI networks of these 37 dysregulated splicing factors. (C, D) The first and second cluster of these splicing factors selected using MCODE app in Cytoscape. (E) Correlation between the firster cluster splicing factors and dysregulated transcripts involved in significant AS events. (F) Correlation between the second cluster splicing factors and dysregulated transcripts involved in significant AS events.

Figure 6. Analysis of upstream splicing factors. (A) volcano plot of dysregulated splicing factors. (B) PPI networks of these 37 dysregulated splicing factors. (C, D) The first and second cluster of these splicing factors selected using MCODE app in Cytoscape. (E) Correlation between the firster cluster splicing factors and dysregulated transcripts involved in significant AS events. (F) Correlation between the second cluster splicing factors and dysregulated transcripts involved in significant AS events.

Table 4. Dysregulated transcripts of splicing factors.

GeneTranscriptlogFCP.ValueChange
DHX32ENSMUST000000636691.3833750.000237UP
AQRENSMUST00000102543−1.48030.000179DOWN
POLR2JENSMUST00000111127−1.116410.008073DOWN
SNRNP35ENSMUST000001114532.0047110.001256UP
HNRNPH3ENSMUST000001198140.9864230.045196UP
LSM11ENSMUST00000129820−0.632950.031731DOWN
POLR2EENSMUST00000131743−0.813050.031261DOWN
DEKENSMUST000001384010.8655510.025996UP
LGALS3ENSMUST000001464682.0613784.77E-07UP
NONOENSMUST000001483120.8425660.049619UP
PRPF40BENSMUST00000150641−0.684160.03879DOWN
SNRPBENSMUST00000150835−0.747670.032351DOWN
PCF11ENSMUST000001511770.8139640.041743UP
THOC2ENSMUST00000151454−1.256720.00612DOWN
POLR2HENSMUST00000161038−1.0310.024872DOWN
UBL5ENSMUST000001618821.0787130.020229UP
TNPO3ENSMUST00000164325−0.802540.047883DOWN
WACENSMUST00000171042−1.051790.035422DOWN
RBM5ENSMUST00000182304−0.734680.005779DOWN
SRRM2ENSMUST000001860450.6181580.014162UP
SF3B1ENSMUST000001890510.6990370.035869UP
PTBP2ENSMUST00000196343−1.065510.005463DOWN
SRPK2ENSMUST000001976220.680850.031996UP
HNRNPUL1ENSMUST00000206260−0.877320.047941DOWN
SNRNP70ENSMUST00000209858−1.003680.013086DOWN
SF3B3ENSMUST000002125150.9261180.043761UP
CCAR1ENSMUST000002184370.6683090.029909UP
CPSF1ENSMUST000002308220.9824230.041531UP
KHDRBS3ENSMUST00000230847−0.642570.006404DOWN
TCERG1ENSMUST000002376020.6580580.017652UP

Discussion

Aβ plaque is a hallmark of AD. Aβ is produced by sequential cleavage of APP, and APP transgenic mice have been widely used ad AD model [13, 14]. In this study we explored the AS events in hippocampal microglia sorted by CD45 and CD11b in APP and Ntg mice. All five types of AS events were detected, and genes involved different AS events showed AS type specific GO enrichment. Aberrant AS have been widely recorded in AD patients and animal models [10, 11]. We for the first time, investigated cell type specific AS events in AD.

Consistent with previous studies [15, 16], SE was the most abundant AS events (68.42%), leading to diverse protein isoforms encoded by a single gene. A3SS (11.58%) and RI (10.53%) showed similar AS event counts. GO analysis of genes involved in significant SE events were enriched in regulation of autophagy (Figure 2A, 2B), including Eif2ak4 and Tpcn2. Eif2ak4 phosphorylates Eif2a in response to various stress stimuli including amino acid starvation. Phosphorylation of Eif2a subsequently promotes translation of activating transcription factor 4 (Atf4). Atf4 is a key autophagy related gene. Atf4 binds to promoters of numerous autophagy genes including Becn1, Atg7, and Sqstm1, and this subsequently increased transcription of these genes. Therefore, activation of Eif2ak4-Eif2a-Atf4 pathway triggers autophagy [17]. Here, we found that Eif2ak4 tended to generate a longer transcript in AD, the function of this longer isoform may deserve further functional studies. Tpcn2 is a lysosomal non-selective Na+/Ca2+ channel, Tpcn2 suppresses the fusion between autophagosome and lysosome, leading to the accumulation of autophagosomes in cancer cells [18]. In this study, we found that Tpcn2 tended to produce a shorter transcript in AD.

A3SS occurred due to alternative acceptor sites, and the PSI indicates inclusion of the longer exon in A3SS events. GO enrichment revealed that genes with significant A3SS events were enriched in actin polymerization and depolymerization, including Mical1 and Add1. Tau accumulation is clearly linked to AD pathogenesis. Mical1 was identified as tau interacting protein via proteomic approach. Mical1 changed the interaction properties of tau and potentiated tau toxicity, mediated by Mical1’s redox activity on tau Cys322 [19]. Moreover, the protein level of Mical1 was increased in AD patients, and considered to be a potential biomarker of tauopathies [19]. Add1 plays essential role in dendritic morphology. Zheng et al. reported that loss of miR-135a-5p expression increased Rock2 activity and phosphorylation of Add1 at Ser726, leading to dendritic abnormalities and memory impairments in AD [20].

Most mRNA derived from RI event may be degraded via NMD, however, some mRNAs could still be translated [11]. Here, we found that genes spliced by RI were enriched in synaptic plasticity including axonogenesis, neurotransmitter secretion, neuron projection and axon guidance (Figure 4A, 4B). Gpc2 tended to harbor an intron in APP group (Figure 4C). Gpc2 has been considered as novel AD risk genes which determine the microglia response to Aβ but not to tau pathology [21].

Moreover, we found that AS events mainly cause dysregulation of genes in transcript level rather than gene expression level (Figure 5C, 5D). Among the 90 genes involved in significant AS event, only 2 genes (Pld3, Dtwd2) were remarkably dysregulated on gene level. However, on transcript level, 26 transcripts refer to 22 genes involved in significant AS events. Rbm3 and Azi2 harbored transcripts with opposite changes in AD. In addition, AS event level was consistent with corresponding transcripts in Azi2 genes (Figure 5E5G), without effect on gene level (Figure 5H). Moreover, 17 out of the 26 transcripts, whose host genes with significant AS events, were involved in SE event, 6 transcripts were involved in RI event (Table 3). These results suggest that SE and RI events may lead to transcript level dysregulation of different genes in hippocampal microglia in AD. We have also performed GO enrichment analysis of the 17 transcripts with SE event, it seems that these transcripts were not associated with the phenotypes of hippocampal microglia (data were not shown).

The upstream SFs were also explored. 37 significantly dysregulated SF transcripts refer to 30 SFs were identified (Figure 6A). Two main clusters were selected from SFs PPI networks, and correlation analysis was performed between these SFs and transcripts involved in AS events. Several SFs showed significantly positive or negative correlation with these transcripts (Figure 6E, 6F). These results suggest that dysregulated SFs may govern the AS events of critical AD pathogenesis related genes described here. We have searched several splicing factor databases to explore AS type-specific preferences of these SFs, including SpliceAid-F (http://www.caspur.it/SpliceAidF/), SpliceAid 2 (http://193.206.120.249/splicing_tissue.html), RBPmap, and MiasDB (http://47.88.84.236/Miasdb/index.php). However, SFs preferences for different five basic modes of AS have not been identified. AS type-specific preferences of SFs need further investigation.

In conclusion, we described hippocampal microglia AS in AD. Autophagy related genes were mainly spliced in SE events, actin depolymerization genes were spliced in A3SS events, while synaptic plasticity related genes were mainly spliced in RI pattern. Dysregulation of autophagy, actin depolymerization, and synaptic plasticity have been widely documented in AD. Our results provide a novel therapeutic direction based on AS regulation. These significant AS events may be regulated by dysregulated splicing factors in AD. Our study provides novel pathological mechanisms in the pathogenesis of AD.

Materials and Methods

Dataset and reference genome

GSE171195 was deposited in GEO (https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE171195), and the raw data were downloaded from ENA database (https://www.ebi.ac.uk/ena/browser/view/PRJNA718619) using axel (v2.17.5) and checked using md5sum (v8.30). GSE171195 performed RNA sequencing of hippocampal microglia cells from APP and Ntg mice. The gtf, cDNA fasta, and DNA fasta files were downloaded from ENSEMBL database as reference.

Reads alignment

FastQC (v0.11.9) and multiqc (v1.11) were used for quality control. Trim-galore (v0.6.7) was used for adaptors and low-quality trimming with parameters “trim_galore --phred33 -q 20 --length 36 --stringency 3 --fastqc --max_n 3”. STAR (v 2.7.10a) was used to align the clean data to the reference genome downloaded from ENSEMBL [22], with parameters “STAR --runThreadN 6 --readFilesCommand zcat --outSAMtype BAM SortedByCoordinate --outBAMsortingThreadN 6”.

Gene/transcript expression matrix

The aligned and sorted bam files were proceeded to generate gene expression matrix with featureCounts (v2.0.1) [23], with parameters “featureCounts -T 8 -p -t exon -g gene_id”. Clean fastq files after data trimming were used to generate transcript expression matrix with salmon (v1.0.4) [24].

Differential expressed gene/transcript analysis

The gene and transcript expression matrices were used for differential expression analysis using limma R package (v3.50.0) [25]. The cutoff values for p value and logFC were 0.5 and 1.5 respectively.

Alternative splicing analysis

The sorted bam files were used for AS analysis with rMATS (v4.1.2) [26] and plotted using rmats2sashimiplot (v2.0.4), with parameters “rmats.py --b1. /b1.txt --b2. /b2.txt --gtf. /Mus_musculus.GRCm39.105.gtf -t single --readLength 100 --nthread 30 --od. /output --tmp. /tmp_output”. Significant AS events were considered if average coverage >5 and delta percent spliced in (ΔPSI) >0.1. Statistics of AS events was performed using maser (v1.12.1) R package. The code for the above methods were similar with our previous codes deposited Github (https://github.com/tjhwangxiong/AD-GSE132177-Alternative-Splicing-pipeline).

Gene ontology enrichment analysis

Gene ontology (GO) enrichment analysis was performed with clusterProfiler (v4.2.2) [27], and GOplot (v1.0.2) [28] R packages. The p value < 0.05 was considered as significant GO terms.

Protein-protein interaction network analysis

The protein-protein interaction (PPI) network analysis was performed using String (https://cn.string-db.org/) and Cytoscape (v3.8.2) [29]. The MCODE app was used to build sub clusters with default settings. The first and second clusters of significantly dysregulated splicing factor transcripts were selected for further transcript correlation analysis.

Transcript correlation analysis

The correlation between significantly dysregulated splicing factor transcripts and significantly dysregulated transcripts whose host genes harbored significant AS events was performed using ggcorrplot R package (v0.1.3). The correlation and p values were calculated. Only correlation results with p value < 0.05 were presented.

Data availability statement

The GSE171195 dataset was deposited in GEO database: https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE171195. Other data were included in the article or supplementary files.

Abbreviations

AS: alternative splicing; 5′SS: 5′ splice site; 3′SS: 3′ splice site; A3SS: alternative 3′SS; SE: skipped exon; RI: retained intron; A5SS: alternative 5′SS; MXE: mutually exclusive exons; AD: Alzheimer’s disease; PSI: percent spliced in; GO: gene ontology; PCA: principal component analysis; NMD: nonsense-mediated mRNA decay.

Author Contributions

Data curation, Y.L., J.X. and L.T.; Funding acquisition, X.W.; Supervision, L.C.; Writing-review and editing, X.W. All authors have read and agreed to the published version of the manuscript.

Acknowledgments

We thank Dr. Jianming Zeng (University of Macau), and all the members of his bioinformatics team, biotrainee, for generously sharing their experience and codes.

Conflicts of Interest

The authors declare no conflicts of interest related to this study.

Funding

This work was supported by Tongji Hospital (HUST) Foundation for Excellent Young Scientist (Grant No. 2020YQ01-11).

References

  • 1. Matlin AJ, Clark F, Smith CW. Understanding alternative splicing: towards a cellular code. Nat Rev Mol Cell Biol. 2005; 6:386–98. https://doi.org/10.1038/nrm1645 [PubMed]
  • 2. Baralle FE, Giudice J. Alternative splicing as a regulator of development and tissue identity. Nat Rev Mol Cell Biol. 2017; 18:437–51. https://doi.org/10.1038/nrm.2017.27 [PubMed]
  • 3. Murphy AJ, Li AH, Li P, Sun H. Therapeutic Targeting of Alternative Splicing: A New Frontier in Cancer Treatment. Front Oncol. 2022; 12:868664. https://doi.org/10.3389/fonc.2022.868664 [PubMed]
  • 4. Liu Z, Rabadan R. Computing the Role of Alternative Splicing in Cancer. Trends Cancer. 2021; 7:347–58. https://doi.org/10.1016/j.trecan.2020.12.015 [PubMed]
  • 5. Zhang Y, Qian J, Gu C, Yang Y. Alternative splicing and cancer: a systematic review. Signal Transduct Target Ther. 2021; 6:78. https://doi.org/10.1038/s41392-021-00486-7 [PubMed]
  • 6. Sengupta U, Kayed R. Amyloid β, Tau, and α-Synuclein aggregates in the pathogenesis, prognosis, and therapeutics for neurodegenerative diseases. Prog Neurobiol. 2022; 214:102270. https://doi.org/10.1016/j.pneurobio.2022.102270 [PubMed]
  • 7. Zhong L, Chen XF. The Emerging Roles and Therapeutic Potential of Soluble TREM2 in Alzheimer's Disease. Front Aging Neurosci. 2019; 11:328. https://doi.org/10.3389/fnagi.2019.00328 [PubMed]
  • 8. Han S, Na Y, Koh I, Nho K, Lee Y. Alternative Splicing Regulation of Low-Frequency Genetic Variants in Exon 2 of TREM2 in Alzheimer's Disease by Splicing-Based Aggregation. Int J Mol Sci. 2021; 22:9865. https://doi.org/10.3390/ijms22189865 [PubMed]
  • 9. Zhang Q, Zhang J, Ye J, Li X, Liu H, Ma X, Wang C, He K, Zhang W, Yuan J, Zhao Y, Xu H, Liu Q. Nuclear speckle specific hnRNP D-like prevents age- and AD-related cognitive decline by modulating RNA splicing. Mol Neurodegener. 2021; 16:66. https://doi.org/10.1186/s13024-021-00485-w [PubMed]
  • 10. Yang M, Ke Y, Kim P, Zhou X. ExonSkipAD provides the functional genomic landscape of exon skipping events in Alzheimer's disease. Brief Bioinform. 2021; 22:bbaa438. https://doi.org/10.1093/bib/bbaa438 [PubMed]
  • 11. Li HD, Funk CC, McFarland K, Dammer EB, Allen M, Carrasquillo MM, Levites Y, Chakrabarty P, Burgess JD, Wang X, Dickson D, Seyfried NT, Duong DM, et al. Integrative functional genomic analysis of intron retention in human and mouse brain with Alzheimer's disease. Alzheimers Dement. 2021; 17:984–1004. https://doi.org/10.1002/alz.12254 [PubMed]
  • 12. Sveen A, Agesen TH, Nesbakken A, Rognum TO, Lothe RA, Skotheim RI. Transcriptome instability in colorectal cancer identified by exon microarray analyses: Associations with splicing factor expression levels and patient survival. Genome Med. 2011; 3:32. https://doi.org/10.1186/gm248 [PubMed]
  • 13. Saito T, Matsuba Y, Mihira N, Takano J, Nilsson P, Itohara S, Iwata N, Saido TC. Single App knock-in mouse models of Alzheimer's disease. Nat Neurosci. 2014; 17:661–3. https://doi.org/10.1038/nn.3697 [PubMed]
  • 14. Esquerda-Canals G, Montoliu-Gaya L, Güell-Bosch J, Villegas S. Mouse Models of Alzheimer's Disease. J Alzheimers Dis. 2017; 57:1171–83. https://doi.org/10.3233/JAD-170045 [PubMed]
  • 15. Florea L, Song L, Salzberg SL. Thousands of exon skipping events differentiate among splicing patterns in sixteen human tissues. F1000Res. 2013; 2:188. https://doi.org/10.12688/f1000research.2-188.v2 [PubMed]
  • 16. Kim P, Yang M, Yiya K, Zhao W, Zhou X. ExonSkipDB: functional annotation of exon skipping event in human. Nucleic Acids Res. 2020; 48:D896–907. https://doi.org/10.1093/nar/gkz917 [PubMed]
  • 17. Bretin A, Carrière J, Dalmasso G, Bergougnoux A, B'chir W, Maurin AC, Müller S, Seibold F, Barnich N, Bruhat A, Darfeuille-Michaud A, Nguyen HT. Activation of the EIF2AK4-EIF2A/eIF2α-ATF4 pathway triggers autophagy response to Crohn disease-associated adherent-invasive Escherichia coli infection. Autophagy. 2016; 12:770–83. https://doi.org/10.1080/15548627.2016.1156823 [PubMed]
  • 18. Sun W, Yue J. TPC2 mediates autophagy progression and extracellular vesicle secretion in cancer cells. Exp Cell Res. 2018; 370:478–89. https://doi.org/10.1016/j.yexcr.2018.07.013 [PubMed]
  • 19. Prifti E, Tsakiri EN, Vourkou E, Stamatakis G, Samiotaki M, Skoulakis EMC, Papanikolopoulou K. Mical modulates Tau toxicity via cysteine oxidation in vivo. Acta Neuropathol Commun. 2022; 10:44. https://doi.org/10.1186/s40478-022-01348-1 [PubMed]
  • 20. Zheng K, Hu F, Zhou Y, Zhang J, Zheng J, Lai C, Xiong W, Cui K, Hu YZ, Han ZT, Zhang HH, Chen JG, Man HY, et al. miR-135a-5p mediates memory and synaptic impairments via the Rock2/Adducin1 signaling pathway in a mouse model of Alzheimer's disease. Nat Commun. 2021; 12:1903. https://doi.org/10.1038/s41467-021-22196-y [PubMed]
  • 21. Sierksma A, Lu A, Mancuso R, Fattorelli N, Thrupp N, Salta E, Zoco J, Blum D, Buée L, De Strooper B, Fiers M. Novel Alzheimer risk genes determine the microglia response to amyloid-β but not to TAU pathology. EMBO Mol Med. 2020; 12:e10606. https://doi.org/10.15252/emmm.201910606 [PubMed]
  • 22. Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, Gingeras TR. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013; 29:15–21. https://doi.org/10.1093/bioinformatics/bts635 [PubMed]
  • 23. Liao Y, Smyth GK, Shi W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics. 2014; 30:923–30. https://doi.org/10.1093/bioinformatics/btt656 [PubMed]
  • 24. Patro R, Duggal G, Love MI, Irizarry RA, Kingsford C. Salmon provides fast and bias-aware quantification of transcript expression. Nat Methods. 2017; 14:417–9. https://doi.org/10.1038/nmeth.4197 [PubMed]
  • 25. Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015; 43:e47. https://doi.org/10.1093/nar/gkv007 [PubMed]
  • 26. Shen S, Park JW, Lu ZX, Lin L, Henry MD, Wu YN, Zhou Q, Xing Y. rMATS: robust and flexible detection of differential alternative splicing from replicate RNA-Seq data. Proc Natl Acad Sci U S A. 2014; 111:E5593–601. https://doi.org/10.1073/pnas.1419161111 [PubMed]
  • 27. Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, Fu X, Liu S, Bo X, Yu G. clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Camb). 2021; 2:100141. https://doi.org/10.1016/j.xinn.2021.100141 [PubMed]
  • 28. Walter W, Sánchez-Cabo F, Ricote M. GOplot: an R package for visually combining expression data with functional analysis. Bioinformatics. 2015; 31:2912–4. https://doi.org/10.1093/bioinformatics/btv300 [PubMed]
  • 29. Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003; 13:2498–504. https://doi.org/10.1101/gr.1239303 [PubMed]