Skip to main content

Integrated identification of key genes and pathways in Alzheimer’s disease via comprehensive bioinformatical analyses

Abstract

Background

Alzheimer’s disease (AD) is known to be caused by multiple factors, meanwhile the pathogenic mechanism and development of AD associate closely with genetic factors. Existing understanding of the molecular mechanisms underlying AD remains incomplete.

Methods

Gene expression data (GSE48350) derived from post-modern brain was extracted from the Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) were derived from hippocampus and entorhinal cortex regions between AD patients and healthy controls and detected via Morpheus. Functional enrichment analyses, including Gene Ontology (GO) and pathway analyses of DEGs, were performed via Cytoscape and followed by the construction of protein-protein interaction (PPI) network. Hub proteins were screened using the criteria: nodes degree≥10 (for hippocampus tissues) and ≥ 8 (for entorhinal cortex tissues). Molecular Complex Detection (MCODE) was used to filtrate the important clusters. University of California Santa Cruz (UCSC) and the database of RNA-binding protein specificities (RBPDB) were employed to identify the RNA-binding proteins of the long non-coding RNA (lncRNA).

Results

251 & 74 genes were identified as DEGs, which consisted of 56 & 16 up-regulated genes and 195 & 58 down-regulated genes in hippocampus and entorhinal cortex, respectively. Biological analyses demonstrated that the biological processes and pathways related to memory, transmembrane transport, synaptic transmission, neuron survival, drug metabolism, ion homeostasis and signal transduction were enriched in these genes. 11 genes were identified as hub genes in hippocampus and entorhinal cortex, and 3 hub genes were identified as the novel candidates involved in the pathology of AD. Furthermore, 3 lncRNAs were filtrated, whose binding proteins were closely associated with AD.

Conclusions

Through GO enrichment analyses, pathway analyses and PPI analyses, we showed a comprehensive interpretation of the pathogenesis of AD at a systematic biology level, and 3 novel candidate genes and 3 lncRNAs were identified as novel and potential candidates participating in the pathology of AD. The results of this study could supply integrated insights for understanding the pathogenic mechanism underlying AD and potential novel therapeutic targets.

Background

Alzheimer’s disease (AD) is recognized as the most common neurodegenerative disease and a typical hippocampal amnesia, and also one of the dominating deadly disease affecting elderly population. The disease is characterized by the extracellular senile plaques formed by amyloid-β (Aβ) peptides, intracellular neurofibrillary tangles (NFTs), and also structure and function changes of brain regions related to memory [1,2,3]. It is well known that AD has complex multiple pathogenic factors, such as genetic factor, environmental factor, immunological factor, head injuries, depression, or hypertension [4,5,6,7,8]. Among these factors, genetic factors are estimated to attribute about 70% to the risk for AD [9]. Dominant mutations of genes encoding APP (amyloid precursor protein), PSEN1 (presenilin 1), and PSEN2 (presenilin 2), which enhanced generation and aggregation of Aβ, were included in the established genetic causes of AD [10]. However, APP, PSEN1 and PSEN2 are only partially accountable for the pathogenic mechanism of AD patients [11, 12]. Besides, genetic analyses have demonstrated that, individual differences of AD could be resulted from multiple genes and their variants, which exert various biological functions in coordination to enhance the risk of the disease [13,14,15]. Except for identifying mechanisms involved in the AD pathogenesis, comprehensive analyses of potential candidate genes could suggest novel potential strategies to predictive or diagnostic test for AD.

Hub genes, regulatory transcription factors and microRNAs in the entorhinal cortex tissues of mid-stage AD cases have been identified via analyzing the database of GSE4757 and therapeutic targets or biomarkers of the AD were demonstrated in previous study [16]. Multiple methods were employed in the identification of potential molecules targets and drug candidates to AD, and hub genes like ZFHX3, ErbB2, ErbB4, OCT3, MIF, CDK13, GPI and so forth were found in the analyses of current datasets, such as GSE48350, GSE36980, GSE5281, and so forth [17,18,19,20,21,22,23]. Whereas, the remarkable and integrated details of key candidate genes and pathways related with the pathogenesis of AD are still incomplete. Furthermore, it is well documented that long non-coding RNAs (lncRNAs) play vital roles in the regulation of gene expression epigenetic, transcriptional, and posttranscriptional levels [24,25,26], and only several lncRNAs have been validated to be involved in the pathogenesis of AD [27,28,29,30]. Given that sufficiently illuminating human lncRNA-AD associations have great potential benefit to diagnosis, prevention, treatment, and prognosis of AD, it is an urgent task to find novel connections between lncRNA and AD.

In the present study, we implemented integrated analyses of genes involved in AD from the information filtration of the database, GSE48350 [31,32,33]. We employed Morpheus, an online tool, to identified differentially expressed genes (DEGs). Then biological enrichment analyses were conducted to detect the remarkable functional terms and analyzed the reciprocities among the biological pathways enriched by pathway analyses methods. Moreover, a protein network specific in AD was speculated and evaluated in the background of the human protein-protein interaction (PPI) network. 3 novel genes and 3 lncRNAs were differently expressed in the tissues of hippocampus and/or entorhinal cortex between AD patients and normal ones were identified as novel and potential candidates of AD pathology, and binding proteins of these lncRNAs closely associate with pathogenic mechanism of AD. The results of the present study should supply ponderable hints for understanding the pathogenesis molecular mechanisms of AD from a standpoint of systems biology.

Results

Identification of DEGs

The gene expression profile and sample information of post-mortem brain tissue samples of AD patients and normal people of GSE48350 were obtained from National Center of Biotechnology Information-Gene Expression Omnibus (NCBI-GEO) and ArrayExpress database, respectively, which are free databases of microarray/gene profile and next-generation sequencing. There were total 253 samples in this database, including microarray data from normal controls and AD cases aged 20–99 years, from 4 brain regions: hippocampus, entorhinal cortex, superior frontal cortex, and post-central gyrus. We analyzed the differences of gene expression between 18 AD samples (69–99 years old) and age-matched 24 normal samples of hippocampus tissues, and 15 AD samples (69–99 years old) and age-matched 17 normal samples of entorhinal cortex tissues in the present study (Additional file 1: Table S1). Employing Morpheus software and using p < 0.05 and |log2FC| ≥ 1 (FC, fold change) as cut-off criterion, 251 genes (56 up-regulated and 195 down-regulated genes) and 74 genes (16 up-regulated and 58 down-regulated genes) were identified as DEGs in the AD samples compared with the normal ones in the tissues of hippocampus and entorhinal cortex, respectively (Table 1 and Additional file 2: Table S2).

Table 1 DEGs were identified from the dataset

Gene ontology (GO) enrichment analyses of DEGs

GO analyses for the DEGs after gene integration were performed via Cytoscape and its plugs, Cluego and Cluepedia. 86 of the 251 DEGs from hippocampus tissues were mapped to 34 different biological processes (Fig. 1a), of which prominent examples are memory 17.65%, response to anesthetic 14.71%, chemical synaptic transmission 11.76% and cellular potassium ion transport 11.76% and neuropilin signaling pathway 11.76% (Fig. 1b and Additional file 3: Table S3). 54 of the 251 DEGs from hippocampus tissues were mapped to 23 different cellular components (Fig. 1c), of which prominent examples are integral component of synaptic membrane 39.13%, leading edge membrane 21.74% (Fig. 1d and Additional file 4: Table S4). 44 of the 251 DEGs from hippocampus tissues were mapped to 19 different molecular functions (Fig. 1e), of which prominent examples are potassium ion transmembrane transporter activity 47.37%, dicarboxylic acid transmembrane transporter activity 15.79% (Fig. 1f and Additional file 5: Table S5). 17 of the 74 DEGs from entorhinal cortex tissues were mapped to 20 different biological processes (Fig. 1g), of which prominent examples are sodium ion homeostasis 45%, positive regulation of muscle contraction 15% and endoderm formation 15% (Fig. 1h and Additional file 6: Table S6).

Fig. 1
figure 1

GO analyses of DEGs. 86 DEGs from hippocampus tissues were mapped to 34 different biological processes (a)(b). a Group information of biological processes. b Percentages of biological processes terms per group. 54 DEGs from hippocampus tissues were mapped to 23 different cellular components (c)(d). c Group information of cellular components. d Percentages of cellular components terms per group. 44 DEGs from hippocampus tissues were mapped to 19 different molecular functions (e)(f). e Group information of molecular functions. f Percentages of molecular functions terms per group. 17 DEGs from entorhinal cortex tissues were mapped to 20 different biological processes (g)(h). g Group information of biological processes. h Percentages of biological processes terms per group

Pathway enrichment analyses of DEGs

In all, pathway enrichment analyses of DEGs were classified by the Kyoto Encyclopedia of Genes and Genomes (KEGG), Reactome and Wikipathway databases, respectively, using p < 0.05 as cut-off value. The different databases provided similar information with the majority of AD-related proteins acting in 19 major pathways (13 from hippocampus and 6 from entorhinal cortex), mainly related transmembrane transportation, drug reactions, synapses function, ion homeostasis, neurogenesis and signal transduction (Additional file 7: Table S7).

PPI network analyses and module analyses

Using the Search Tool for the Retrieval of Interacting Genes database (STRING) online database and Cytoscape software, total of 135 DEGs (26 up-regulated and 109 down-regulated genes) of the 251 commonly altered DEGs from hippocampus were screened into the DEGs PPI network, containing 135 nodes and 221 edges (Fig. 2a), and 116 of the 251 DEGs did not fall into the DEGs PPI network. Based on the STRING database, we made the PPI network of a total of 135 nodes and 221 protein pairs was obtained with a combined score > 0.4. As shown in Fig. 2a, the majority of the nodes in the network were down-regulated DEGs in AD samples. Among the 135 nodes, 9 central node genes were identified with the filtering of degree≥10 criteria (i.e., each node had more than 10 connections/interactions) as top 9 hub genes, which were CDC42, BDNF, TH, PDYN, VEGFA, CALB, CD44, TAC1 and CACNA1A (Fig. 2b). Figure 2b presents these 9 genes and their first neighbor genes.

Fig. 2
figure 2

PPI networks and clusters of DEGs from hippocampus. a PPI networks of all 135 proteins. b Hub genes and their neighbor genes. c Module 1 from the PPI network. d Module 2 from the PPI network

In total, 2 modules (Modules 1 and 2) with score > 3 were detected by Cytoscape plug-in Molecular Complex Detection (MCODE) (Fig. 2c and d). KEGG pathway enrichment analyses showed that Module 1 consisted of 4 nodes and 5 edges, which are mainly associated with Endocytosis, Rap1 signaling pathway and Ras signaling pathway (Table 2), and that Module 2 consisted of 10 nodes and 14 edges, which are mainly associated with neuroactive ligand-receptor interaction, and cocaine addiction (Table 3).

Table 2 Pathway enrichment in Module 1
Table 3 Pathway enrichment in Module 2

Similarly, total of 32 DEGs (6 up-regulated and 26 down-regulated genes) of the 74 commonly altered DEGs from entorhinal cortex were screened into the DEGs PPI network, containing 32 nodes and 41 edges (Fig. 3a), and 42 of the 74 DEGs did not fall into the DEGs PPI network. As shown in Fig. 3a, the majority of the nodes in the network were down-regulated DEGs in AD samples. Among the 32 nodes, 2 central node genes were identified with the filtering of degree≥8 criteria (i.e., each node had more than 8 connections/interactions) as top 2 hub genes, which were OXT and TAC1 (Fig. 3b). Figure 3b presents these 2 genes and their first neighbor genes.

Fig. 3
figure 3

PPI networks and clusters of DEGs from entorhinal cortex. a PPI networks of all 32 proteins. b Hub genes and their neighbor genes. c Module 3 from the PPI network

In total, 1 module (Modules 3) with score > 3 was detected by MCODE (Fig. 3c). KEGG pathway enrichment analyses showed that Module 3 consisted of 5 nodes and 10 edges, which are mainly associated with signal transduction (including multiple receptors) (Table 4).

Table 4 Pathway enrichment in Module 3

Identification of lncRNAs and analysis of binding proteins

21 mutual DEGs were discovered through the tool of venn by employing Funrich software, and among which 1 lncRNA, linc00622, was identified as differently expressed both in the tissues of hippocampus and entorhinal cortex between AD patients and normal controls (Fig. 4a). Figure 4b and c show the relative expressed values of linc00622 in the tissues of hippocampus and entorhinal cortex. Linc00282 and linc00960 were differently expressed in the tissues of hippocampus and entorhinal cortex, respectively. After using the online tools, University of Califorina Santa Cruz (UCSC) and the Database of RNA-binding protein specificities (RBPDB), we obtained the RNA-binding proteins lists of linc00662, linc00282 and linc00960. Then, 14 mutual RNA-binding proteins were found to be shared by these 3 lncRNAs via the tool of venn (Fig. 4d). The biofunctions of these RNA-binding proteins were summarized in the Table 5.

Fig. 4
figure 4

LncRNAs identification and filtration of mutual binding proteins. a Mutual DEGs between hippocampus and entorhinal cortex. b Relative expression of linc00622 in hippocampus. c Relative expression of linc00622 in entorhinal cortex. d Mutual RNA-binding proteins among linc00622, linc00282 and linc00960

Table 5 Information of RNA-binding proteins of lncRNAs

Discussion

In general, AD is a genetically complex neurodegenerative disease and is characterized by the presence of extracellular deposition of senile plaques, intracellular NFTs and loss of neuron and synapses [50, 51]. AD affects patients’ living quality and is detrimental to their life, and further imposes a considerable burden on their families and the whole society. However, there are rare effective therapies for AD patients nowadays; it is urgent to develop novel perspectives to improve treatment outcomes [52].

We selected GSE48350 database, which contains microarray data from AD cases (aged 20–99 years) and age matched normal controls, from 4 brain regions: hippocampus, entorhinal cortex, superior frontal cortex, and post-central gyrus. Previous study has demonstrated that frontal cortical dysfunction contributed a significant extent to cognitive deficits and memory loss, which was considered as the late characteristic of AD [53]; meanwhile, AD has been widely considered as an early amnesic syndrome of hippocampal type, which on behalf of the most significant clinic feature for the diagnosis of AD [54,55,56], and we believed that the data between age-matched AD patients and normal controls were more convictive. Besides, entorhinal cortex is also considered as a vital brain region in characterizing AD, and aberrant changes of entorhinal cortex happen before hippocampus in the pathological mechanism of AD [57,58,59]. Therefore, we analyzed the data from hippocampus and entorhinal cortex tissues between aged AD cases from 69 to 99 years and age matched normal controls. We employed several types of tools to recognize critical molecular terms and mechanisms involved in AD.

We firstly used Morpheus to filtrate the DEGs from post-mortem samples between AD patients and normal controls using p < 0.05 and |log2FC| ≥ 1 as the criteria, and then we obtained 251 DEGs including 56 up-regulated genes and 195 down-regulated genes in hippocampus tissues, and 74 DEGs including 16 up-regulated genes and 58 down-regulated genes in entorhinal cortex tissues.

The overrepresented biological processes, cellular components and molecular functions obtained from GO analyses of DEGs from hippocampus and entorhinal cortex tissues may give valuable information about the pathogenic molecular mechanisms of AD. Among the GO terms overrepresented in AD patients, those related to memory-related processes, drug reactions, transmembrane transportation, synaptic transmission and ion homeostasis were included. These results were in accordance with previous studies that complex interrelationships of synaptic depression, cognitive impairment, aberrant drug metabolism, and imbalance of ion homeostasis existed among the nosetiology and development processes of AD [60,61,62,63,64,65]. The multiformity in the biological process of genes involved in AD indicated the complicacy of the disease. Recent studies convinced that ion channels are well-known to be involved in AD pathophysiology, especially potassium ion channel, and is emerging as a new target candidate for AD [66, 67].

Pathway enrichment analyses of DEGs from hippocampus and entorhinal cortex tissues were classified by the Reactome, KEGG and/or Wikipathway databases, respectively. The different databases provided similar information with the majority of AD-related proteins acting in 19 major pathways (13 from hippocampus and 6 from entorhinal cortex), mainly related transmembrane transportation, drug reactions, synapses function, ion homeostasis, neurogenesis and signal transduction, which were consistent with the results of GO analyses of these DEGs and previous works focused on the aetiology of AD [60,61,62,63,64,65, 68]. Besides, in the pathway analyses of 3 modules (2 for DEGs from hippocampus and 1 for entorhinal cortex), it was indicated that multiple signal transduction pathways were involved in the pathological mechanism of AD.

From the results of functional enrichment analyses of DEGs, it can be concluded that synaptic depression, cognitive impairment, aberrant drug metabolism, and imbalance of ion homeostasis participated in the pathology of AD and signaling pathways that regulate these biological phenomena would be the efficient treatment targets for AD.

Among 11 hub genes (9 from hippocampus and 2 from entorhinal cortex) obtained from PPI network analyses in this study, several genes involved in the regulation of cell survival and cell growth, such as CDC42 and VEGFA; several genes involved in the memory, learning, and cognitive functions, such as BDNF, PDYN, CALB, TH, CACNA1A, and OXT; several genes involved in the immune and neuroprotective functions, such as CD44 and TAC1. Detailed information of these genes is as seen-shown in Table 6. Specially, as far as we know, CALB, CACNA1A and OXT were identified as the hub participants in the pathological mechanism of AD for the first time in this study.

Table 6 Detailed information of hub genes

Given multiple biofunctions of human lncRNA, the associations between lncRNA and AD have great potential benefits to understanding the cause of AD. So far, only several lncRNAs, such as BACe1-AS [27], 51A [28], 17A [29], BC200 [95] and so on, have been validated to be involved in the pathogenesis of AD. Identifying potential diagnostic lncRNA biomarkers by employing computational methods is promising in the biomarker filtration for AD. In the present study, among 21 mutual DEGs of hippocampus and entorhinal cortex tissues, 1 lncRNA, linc00622 was identified as differently expressed both in the tissues of hippocampus and entorhinal cortex. Besides, linc00282 and linc00960 were differently expressed in the tissues of hippocampus and entorhinal cortex, respectively. 14 mutual RNA-binding proteins were proved to have close relationship with paroxysm and development of AD. Therefore, linc00622, linc00282 and linc00960 could be considered as novel potential candidates participating in the pathological mechanism of AD.

Conclusions

Combined the results of comprehensive and systematic analyses focusing on the biological functions and interactions of the genes extracted from GSE48350 genome database of AD patients and normal controls, genes mainly related the biological functions of memory, synapse, neuron survival, drug metabolism, ion homeostasis and signal transduction were differently expressed in the hippocampus and entorhinal cortex tissues of AD patients aged from 69 to 99 years and age matched normal controls. Our study should shed some light toward a better understanding of the underlying molecular mechanisms and crucial molecular players of AD, and provide a new viewpoint for researchers with target the cause of the disease, and also these understandings need to be further validated by experiments in the future.

Materials and methods

Microarray data extraction and identification of DEGs

The network-based analyses of AD began with the authentication of microarray gene expression dataset. We downloaded the gene expression profile and sample information of GSE48350 from the public availability repository GEO database (https://www.ncbi.nlm.nih.gov/geo/) [96] from NCBI and European Bioinformatics Institute’s (EBI) ArrayExpress-functional genomics database (https://www.ebi.ac.uk/arrayexpress/) [97]. GSE48350 contains post-mortem brain tissue samples from diseased (patients with AD) and control (normal) conditions. Hippocampus region of brain plays significant role in memory formation, which is necessary in diagnostics since loss of memory and cognitive competence and disorientation are the early signs of AD [56]. In this study, we analyzed 18 AD samples of hippocampus region of post-mortem brain aged from 69 to 99 (mean age 84.33 ± 6.56 years) and 24 age-matched control samples of hippocampus region of post-mortem brain (mean age 82.71 ± 9.47 years), and 15 AD samples of entorhinal cortex region aged from 69 to 99 (mean age 86.47 ± 5.46 years) and 17 age-matched control samples of entorhinal cortex region (mean age 81.65 ± 9.76 years).

Morpheus (https://software.broadinstitute.org/morpheus/) [98] online tool allows researchers to carry out of GEO data to identify DEGs. A gene is defined as a DEG between the patients’ samples and the normal control samples when the p-value is < 0.05 and the FC is at least 2 times higher or lower (|log2FC| ≥ 1).

Functional enrichment analyses for DEGs

Executing functional enrichment analyses for DEGs gives a functional overview of the DEGs through computing the whole conspicuousness of the gene expression. GO comprises biological process, cellular component, and molecular function, providing biological functional interpretation of large lists of genes screened from genomic studies such as microarray and proteomics experiments [99, 100]. KEGG is an encyclopedical database resource consisting of graphical diagrams of biochemical pathways for functional gene and molecules to be integrally analyzed [101, 102]. Reactome, an online bioinformatics resource of pathway information, supplies integrated analysis of the biologic reaction network [103, 104]. WikiPathway provides a database in a curated, machine readable way to analyze and visualize data [105]. Pathway analyses of KEGG, Reactome and Wikipathway were employed to illuminate how DEGs perform function through a certain path.

We selected Functional Enrichment analysis tool (Cytoscape v3.7.0), which is an autocephalous software tool employed mainly for functional enrichment and interaction network analyses of genes and proteins. Cytoscape is open source software who can integrate interaction networks of high-throughput expression data and other molecular states of genes and proteins into a unitive conceptual framework [106]. This software has been widely employed by researchers to study biological domains, the genome, proteome and metabonomics [107,108,109,110]. The functional enrichment analyses for the up-regulated and down-regulated DEGs and pathways were performed via Cytoscape and its plugins, ClueGO v2.5.3 and Cluepedia v1.5.3, using p < 0.05 as the selected criterion in the present study.

Construction of PPI network for DEGs and recognition of hub proteins

PPI was employed to analyze the interrelationship among DEGs, and further illustrate the models of genes which play significant roles in physiological and pathological status. The STRING database (https://www.string-db.org/) [111] supplies information about the predicted and experimental interrelationships of proteins, and helps to assess and integrate PPI, including direct (physical) and indirect (functional) correlations [112, 113]. In this study, the DEGs were mapped into PPI using STRING database v10.5. Then, Cytoscape software was employed to visualize the PPI network. The network module was one of the peculiarities of the protein network and contains peculiar biological importance. The MCODE (v1.5.1) was employed to identify remarkable modules in this PPI network. Degree cutoff = 2, Node Score Cutoff = 0.2, and K-Core = 2 were set as the advanced settings. MCODE was applied to filtrate hub proteins within the PPI network. At last, the enrichment analyses of the DEGs in different modules were also conducted by the STRING database.

Identification of lncRNAs and binding proteins prediction

Mutual DEGs were discovered through the tool of venn by employing Funrich (3.1.3) software. The relative expressed values of linc00622 in the tissues of hippocampus and entorhinal cortex were analyzed by Graphpad Prism (7.0) software. Online tools, UCSC (https://genome.ucsc.edu/index.html) [114, 115] and RBPDB (http://rbpdb.ccbr.utoronto.ca/index.php) [116, 117] were used to obtain the RNA-binding proteins lists of linc00662, linc00282 and linc00960. Then, mutual RNA-binding proteins shared by these 3 lncRNAs were found via the tool of venn.

Availability of data and materials

All data generated or analyzed during this study are included in this published article [and its supplementary information files].

Abbreviations

AD:

Alzheimer’s disease

APP:

amyloid precursor protein

Aβ:

amyloid-β

DEGs:

differentially expressed genes

EBI:

European Bioinformatics Institute

FC:

fold change

GEO:

Gene Expression Omnibus

GO:

Gene Ontology

KEGG:

Kyoto Encyclopedia of Genes and Genomes

LncRNA:

long non-coding RNA

MCODE:

Molecular Complex Detection

NCBI:

National Center of Biotechnology Information

NFTs:

neurofibrillary tangles

PPI:

protein-protein interaction

PSEN1:

presenilin 1

PSEN2:

presenilin 2

RBPDB:

the Database of RNA-binding protein specificities

STRING:

Search Tool for the Retrieval of Interacting Genes

UCSC:

University of California Santa Cruz

References

  1. Bakota L, Brandt R. Tau biology and tau-directed therapies for Alzheimer's disease. Drugs. 2016;76(3):301–13. https://doi.org/10.1007/s40265-015-0529-0.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Forner S, Baglietto-Vargas D, Martini AC, et al. Synaptic impairment in Alzheimer's disease: a dysregulated symphony. Trends Neurosci. 2017;40(6):347–57. https://doi.org/10.1016/j.tins.2017.04.002.

    Article  CAS  PubMed  Google Scholar 

  3. Eimer WA, Vijaya Kumar DK, Navalpur Shanmugam NK, et al. Alzheimer’s disease- associated β-amyloid is rapidly seeded by herpesviridae to protect against brain infection. Neuron. 2018;99(1):56–63.e3. https://doi.org/10.1016/j.neuron.2018.06.030.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Jayaraman A, Pike CJ. Alzheimer’s disease and type 2 diabetes: multiple mechanisms contribute to interactions. Curr Diabetes Rep. 2014;14(4):476. https://doi.org/10.1007/s11892-014-0476-2.

    Article  CAS  Google Scholar 

  5. Reitz C. Genetic diagnosis and prognosis of Alzheimer’s disease: challenges and opportunities. Expert Rev Mol Dia. 2015;15(3):339–48. https://doi.org/10.1586/14737159.2015.1002469.

    Article  CAS  Google Scholar 

  6. Rivera DS, Inestrosa NC, Bozinovic F. On cognitive ecology and the environmental factors that promote Alzheimer disease: lessons from Octodon degus (Rodentia: Octodontidae). Biol Res. 2016;49(1):10. https://doi.org/10.1186/s40659-016-0074-7.

    Article  PubMed  PubMed Central  Google Scholar 

  7. Vijayan M, Reddy PH. Stroke and vascular dementia and Alzheimer’s disease-molecular links. J Alzheimers Dis. 2016;54(2):427–43. https://doi.org/10.3233/JAD-160527.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Tolppanen AM, Taipale H, Hartikainen S. Head or brain injuries and Alzheimer’s disease: a nested case-control register study. Alzheimers Dement. 2017;13(12):1371–9. https://doi.org/10.1016/j.jalz.2017.04.010.

    Article  PubMed  Google Scholar 

  9. Girard H, Potvin O, Nugent S, et al. Faster progression from MCI to probable AD for carriers of a single-nucleotide polymorphism associated with type 2 diabetes. Neurobiol Aging. 2018;64:157.e11–7. https://doi.org/10.1016/j.neurobiolaging.2017.11.013.

    Article  CAS  Google Scholar 

  10. Shao W, Peng D, Wang X. Genetics of Alzheimer’s disease: from pathogenesis to clinical usage. J Clin Neurosci. 2017;45:1–8. https://doi.org/10.1016/j.jocn.2017.06.074.

    Article  CAS  PubMed  Google Scholar 

  11. Cruts M, Theuns J, Van Broeckhoven C. Locus-specific mutation databases for neurodegenerative brain diseases. Hum Mutat. 2012;33(9):1340–4. https://doi.org/10.1002/humu.22117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Guerreiro RJ, Gustafson DR, Hardy J. The genetic architecture of Alzheimer's disease: beyond APP, PSENs and APOE. Neurobiol Aging. 2012;33(3):437–56. https://doi.org/10.1016/j.neurobiolaging.2010.03.025.

    Article  CAS  PubMed  Google Scholar 

  13. Hokama M, Oka S, Leon J, et al. Altered expression of diabetes-related genes in Alzheimer’s disease brains: the hisayama study. Cereb Cortex. 2014;24(9):2476–88. https://doi.org/10.1093/cercor/bht101.

    Article  PubMed  Google Scholar 

  14. Naughton BJ, Duncan FJ, Murrey DA, et al. Blood genome-wide transcriptional profiles reflect broad molecular impairments and strong blood-brain links in Alzheimer’s disease. J Alzheimers Dis. 2015;43(1):93–108. https://doi.org/10.3233/JAD-140606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Stopa EG, Tanis KQ, Miller MC, et al. Comparative transcriptomics of choroid plexus in Alzheimer’s disease, frontotemporal dementia and Huntington’s disease: implications for CSF homeostasis. Fluids Barriers of the CNS. 2018;15(1):18–28. https://doi.org/10.1186/s12987-018-0102-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rahman MR, Islam T, Turanli B, et al. Network-based approach to identify molecular signatures and therapeutic agents in Alzheimer’s disease. Comput Biol Chem. 2019;78:431–9. https://doi.org/10.1016/j.compbiolchem.2018.12.011.

    Article  CAS  PubMed  Google Scholar 

  17. Elkahloun AG, Hafko R, Saavedra JM. An integrative genome-wide transcriptome reveals that candesartan is neuroprotective and a candidate therapeutic for Alzheimer’s disease. Alzheimers Res Ther. 2016;8(1):5. https://doi.org/10.1186/s13195-015-0167-5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Pang X, Zhao Y, Wang J, et al. The bioinformatic analysis of the dysregulated genes and microRNAs in entorhinal cortex, hippocampus, and blood for Alzheimer's disease. Biomed Res Int. 2017;2017:9084507. https://doi.org/10.1155/2017/9084507.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Bae SH, Kim HW, Shin SJ, et al. Decipher reliable biomarkers of brain aging by integrating literature-based evidence with interactome data. Biomed Res Int. 2018;50(4):28. https://doi.org/10.1038/s12276-018-0057-6.

    Article  CAS  Google Scholar 

  20. Kim BY, Lim HS, Kim Y, et al. Evaluation of animal models by comparison with human late-onset Alzheimer’s disease. Mol Neurobiol. 2018;55(12):9234–50. https://doi.org/10.1007/s12035-018-1036-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lanke V, Moolamalla STR, Roy D, et al. Integrative analysis of hippocampus gene expression profiles identifies network alterations in aging and Alzheimer’s disease. Front Aging Neurosci. 2018;10:153. https://doi.org/10.3389/fnagi.2018.00153.

    Article  PubMed  PubMed Central  Google Scholar 

  22. Vargas DM, De Bastiani MA, Zimmer ER, et al. Alzheimer’s disease master regulators analysis: search for potential molecular targets and drug repositioning candidates. Alzheimers Res Ther. 2018;10(1):59. https://doi.org/10.1186/s13195-018-0394-7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Xiang S, Huang Z, Wang T, et al. Condition-specific gene co-expression network mining identifies key pathways and regulators in the brain tissue of Alzheimer's disease patients. BMC Med Genet. 2018;11(Suppl 6):115. https://doi.org/10.1186/s12920-018-0431-1.

    Article  CAS  Google Scholar 

  24. Kopp F, Mendell JT. Functional classification and experimental dissection of long noncoding RNAs. Cell. 2018;172(3):393–407. https://doi.org/10.1016/j.cell.2018.01.011.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Churchman LS. Not just noise: genomics and genetics bring long noncoding RNAs into focus. Mol Cell. 2017;65(1):1–2. https://doi.org/10.1016/j.molcel.2016.12.017.

    Article  CAS  PubMed  Google Scholar 

  26. Carlevaro-Fita J, Johnson R. Global positioning system: understanding long noncoding RNAs through subcellular localization. Mol Cell. 2019;73(5):869–83. https://doi.org/10.1016/j.molcel.2019.02.008.

    Article  CAS  PubMed  Google Scholar 

  27. Faghihi MA, Modarresi F, Khalil AM, et al. Expression of a noncoding RNA is elevated in Alzheimer’s disease and drives rapid feed-forward regulation of beta-secretase. Nat Med. 2008;14(7):723–30. https://doi.org/10.1038/nm1784.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Ciarlo E, Massone S, Penna I, et al. An intronic ncRNA-dependent regulation of SORL1 expression affecting Abeta formation is upregulated in post-mortem Alzheimer’s disease brain samples. Dis Mod Mech. 2013;6(2):424–33. https://doi.org/10.1242/dmm.009761.

    Article  CAS  Google Scholar 

  29. Gavazzo P, Vassalli M, Costa D, et al. Novel ncRNAs transcribed by pol III and elucidation of their functional relevance by biophysical approaches. Front Cell Neurosci. 2013;7:203. https://doi.org/10.3389/fncel.2013.00203.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Iacoangeli A, Bianchi R, Tiedge H. Regulatory RNAs in brain function and disorders. Brain Res. 2010;1338:36–47. https://doi.org/10.1016/j.brainres.2010.03.042.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Berchtold NC and Cotman CW. Alzheimer's disease dataset. Gene Expression Omnibus 2014. https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE48350. Accessed 17 Jun 2019.

  32. Berchtold NC, Cribbs DH, Coleman PD, et al. Gene expression changes in the course of normal brain aging are sexually dimorphic. P Natl Acad Sci USA. 2008;105(40):15605–10. https://doi.org/10.1073/pnas.0806883105.

    Article  Google Scholar 

  33. Berchtold NC, Coleman PD, Cribbs DH, et al. Synaptic genes are extensively downregulated across multiple brain regions in normal human aging and Alzheimer's disease. Neurobiol Aging. 2013;34(6):1653–61. https://doi.org/10.1016/j.neurobiolaging.2012.11.024.

    Article  CAS  PubMed  Google Scholar 

  34. Eom T, Muslimov IA, Tsokas P, et al. Neuronal BC RNAs cooperate with eIF4B to mediate activity-dependent translational control. J Cell Biol. 2014;207(2):237–52. https://doi.org/10.1083/jcb.201401005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Peng Y, Yuan J, Zhang Z, et al. Cytoplasmic poly(a)-binding protein 1 (PABPC1) interacts with the RNA-binding protein hnRNPLL and thereby regulates immunoglobulin secretion in plasma cells. J Biol Chem. 2017;292(29):12285–95. https://doi.org/10.1074/jbc.M117.794834.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Scekic-Zahirovic J, Oussini HE, Mersmann S, et al. Motor neuron intrinsic and extrinsic mechanisms contribute to the pathogenesis of FUS-associated amyotrophic lateral sclerosis. Acta Neuropathol. 2017;133(6):887–906. https://doi.org/10.1007/s00401-017-1687-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Zhang M, Chen D, Xia J, et al. Post-transcriptional regulation of mouse neurogenesis by Pumilio proteins. Genes Dev. 2017;31(13):1354–69. https://doi.org/10.1101/gad.298752.117.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Ehrmann I, Dalgliesh C, Liu Y, et al. The tissue-specific RNA binding protein T-STAR controls regional splicing patterns of neurexin pre-mRNAs in the brain. PLoS Genet. 2013;9(4):e1003474. https://doi.org/10.1371/journal.pgen.1003474.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Yoshimi N, Futamura T, Bergen SE, et al. Cerebrospinal fluid metabolomics identifies a key role of isocitrate dehydrogenase in bipolar disorder: evidence in support of mitochondrial dysfunction hypothesis. Mol Psychiatry. 2016;21(11):1504–10. https://doi.org/10.1038/mp.2015.217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Briata P, Bordo D, Puppo M, et al. Diverse roles of the nucleic acid-binding protein KHSRP in cell differentiation and disease. Wires RNA. 2016;7(2):227–40. https://doi.org/10.1002/wrna.1327.

    Article  CAS  PubMed  Google Scholar 

  41. Roundtree IA, Luo GZ, Zhang Z, et al. YTHDC1 mediates nuclear export of N6-methyladenosine methylated mRNAs. Elife. 2017;6:e31311. https://doi.org/10.7554/eLife.31311.

    Article  PubMed  PubMed Central  Google Scholar 

  42. Tariq A, Garncarz W, Handl C, et al. RNA-interacting proteins act as site-specific repressors of ADAR2-mediated RNA editing and fluctuate upon neuronal stimulation. Nucleic Acids Res. 2013;41(4):2581–93. https://doi.org/10.1093/nar/gks1353.

    Article  CAS  PubMed  Google Scholar 

  43. Shashi V, Xie P, Schoch K, et al. The RBMX gene as a candidate for the Shashi X-linked intellectual disability syndrome. Clin Genet. 2015;88(4):386–90. https://doi.org/10.1111/cge.12511.

    Article  CAS  PubMed  Google Scholar 

  44. Ling IF, Estus S. Role of SFRS13A in low-density lipoprotein receptor splicing. Hum Mutat. 2010;31(6):702–9. https://doi.org/10.1002/humu.21244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Matsumoto Y, Itou J, Sato F, et al. SALL4-KHDRBS3 network enhances stemness by modulating CD44 splicing in basal-like breast cancer. Cancer Med. 2018;7(2):454–62. https://doi.org/10.1002/cam4.1296.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Kraushar ML, Thompson K, Wijeratne HR, et al. Temporally defined neocortical translation and polysome assembly are determined by the RNA-binding protein Hu antigen R. P Natl Acad Sci USA. 2014;111(36):E3815–24. https://doi.org/10.1073/pnas.1408305111.

    Article  CAS  Google Scholar 

  47. Skliris A, Papadaki O, Kafasla P, et al. Neuroprotection requires the functions of the RNA-binding protein HuR. Cell Death Differ. 2015;22(5):703–18. https://doi.org/10.1038/cdd.2014.158.

    Article  CAS  PubMed  Google Scholar 

  48. Meseguer S, Mudduluru G, Escamilla JM, et al. MicroRNAs-10a and -10b contribute to retinoic acid-induced differentiation of neuroblastoma cells and target the alternative splicing regulatory factor SFRS1 (SF2/ASF). J Biol Chem. 2011;286(6):4150–64. https://doi.org/10.1074/jbc.M110.167817.

    Article  CAS  PubMed  Google Scholar 

  49. Kanadia RN, Clark VE, Punzo C, et al. Temporal requirement of the alternative-splicing factor Sfrs1 for the survival of retinal neurons. Development. 2008;135(23):3923–33. https://doi.org/10.1242/dev.024620.

    Article  CAS  PubMed  Google Scholar 

  50. De SB and Karran E. The cellular phase of Alzheimer's disease. Cell. 2016;164(4):603–615. DOI: https://doi.org/10.1016/j.cell.2015.12.056.

    Article  PubMed  Google Scholar 

  51. Verheijen J, Sleegers K. Understanding Alzheimer disease at the interface between genetics and transcriptomics. Trends Genet. 2018;34(6):434–47. https://doi.org/10.1016/j.tig.2018.02.007.

    Article  CAS  PubMed  Google Scholar 

  52. Theleritis C, Siarkos K, Katirtzoglou E, et al. Pharmacological and nonpharmacological treatment for apathy in Alzheimer disease. J Geriatr Psych Neur. 2017;30(1):26–49. https://doi.org/10.1177/0891988716678684.

    Article  Google Scholar 

  53. Xu M, Liu Y, Huang Y, et al. Re-exploring the core genes and modules in the human frontal cortex during chronological aging: insights from network-based analysis of transcriptomic studies. Aging (Albany). 2018;10(10):2816–31. https://doi.org/10.18632/aging.101589.

    Article  Google Scholar 

  54. Marchetti C, Marie H. Hippocampal synaptic plasticity in Alzheimer’s disease: what have we learned so far from transgenic models? Rev Neurosci. 2011;22(4):373–402. https://doi.org/10.1515/RNS.2011.035.

    Article  CAS  PubMed  Google Scholar 

  55. Li WX, Dai SX, Liu JQ, et al. Integrated analysis of Alzheimer’s disease and schizophrenia dataset revealed different expression pattern in learning and memory. J Alzheimers Dis. 2016;51:417–25. https://doi.org/10.3233/JAD-150807.

    Article  CAS  PubMed  Google Scholar 

  56. Evans TE, Adams HHH, Licher S, et al. Subregional volumes of the hippocampus in relation to cognitive function and risk of dementia. NeuroImage. 2018;178:129–35. https://doi.org/10.1016/j.neuroimage.2018.05.041.

    Article  PubMed  Google Scholar 

  57. Meda SA, Koran ME, Pryweller JR, et al. Genetic interactions associated with 12-month atrophy in hippocampus and entorhinal cortex in Alzheimer's disease neuroimaging initiative. Neurobiol Aging. 2013;34(5):1518.e9–1518.e18. https://doi.org/10.1016/j.neurobiolaging.2012.09.020.

    Article  CAS  Google Scholar 

  58. Thangavel R, Kempuraj D, Stolmeier D, et al. Glia maturation factor expression in entorhinal cortex of Alzheimer’s disease brain. Neurochem Res. 2013;38(9):1777–84. https://doi.org/10.1007/s11064-013-1080-6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Fu H, Rodriguez GA, Herman M, et al. Tau pathology induces excitatory neuron loss, grid cell dysfunction, and spatial memory deficits reminiscent of early Alzheimer’s disease. Neuron. 2017;93(3):533–541.e5. https://doi.org/10.1016/j.neuron.2016.12.023.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Ma T, Trinh MA, Wexler AJ, et al. Suppression of eif2α kinases alleviates ad-related synaptic plasticity and spatial memory deficits. Nat Neurosci. 2013;16(9):1299–305. https://doi.org/10.1038/nn.3486.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Seo J, Giusti-Rodríguez P, Zhou Y, et al. Activity-dependent p25 generation regulates synaptic plasticity and Aβ-induced cognitive impairment. Cell. 2014;157(2):486–98. https://doi.org/10.1016/j.cell.2014.01.065.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Toledo JB, Arnold M, Kastenmüuller G, et al. Metabolic network failures in Alzheimer’s disease: a biochemical road map. Alzheimers Dement. 2017;13(9):965–84. https://doi.org/10.1016/j.jalz.2017.01.020.

    Article  PubMed  PubMed Central  Google Scholar 

  63. Zhang Q, Yang C, Liu T, et al. Citalopram restores short-term memory deficit and non-cognitive behaviors in APP/PS1 mice while halting the advance of Alzheimer’s disease-like pathology. Neuropharmacology. 2017;131:475–86. https://doi.org/10.1016/j.neuropharm.2017.12.021.

    Article  CAS  PubMed  Google Scholar 

  64. Tracy TE, Sohn PD, Minami SS, et al. Acetylated tau obstructs KIBRA-mediated signaling in synaptic plasticity and promotes tauopathy-related memory loss. Neuron. 2016;90(2):245–60. https://doi.org/10.1016/j.neuron.2016.03.005.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. de Lores Arnaiz GR, Ordieres MG. Brain Na(+), K(+)-ATPase activity in aging and disease. J Biomed Sci. 2014;10(2):85–102.

    Google Scholar 

  66. Boscia F, Pannaccione A, Ciccone R, et al. The expression and activity of Kv3.4 channel subunits are precociously upregulated in astrocytes exposed to Aβ oligomers and in astrocytes of Alzheimer's disease Tg2576 mice. Neurobiol Aging. 2017;54:187–98. https://doi.org/10.1016/j.neurobiolaging.2017.03.008.

    Article  CAS  PubMed  Google Scholar 

  67. Esmaeili MH, Bahari B, Salari AA. ATP-sensitive potassium-channel inhibitor glibenclamide attenuates HPA axis hyperactivity, depression-and anxiety-related symptoms in a rat model of Alzheimer’s disease. Brain Res Bull. 2018;137:265–76. https://doi.org/10.1016/j.brainresbull.2018.01.001.

    Article  CAS  PubMed  Google Scholar 

  68. Deng PY, Klyachko VA. Increased persistent sodium current causes neuronal hyperexcitability in the entorhinal cortex of fmr1 knockout mice. Cell Rep. 2016;16(12):3157–66. https://doi.org/10.1016/j.celrep.2016.08.046.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Takai Y, Sasaki T, Matozaki T. Small GTP-binding proteins. Physiol Rev. 2001;81(1):153–208. https://doi.org/10.1152/physrev.2001.81.1.153.

    Article  CAS  PubMed  Google Scholar 

  70. Burridge K, Wennerberg K. Rho and Rac take center stage. Cell. 2004;116(2):167–79. https://doi.org/10.1016/s0092-8674(04)00003-0.

    Article  CAS  PubMed  Google Scholar 

  71. Parri M, Chiarugi P. Rac and rho GTPases in cancer cell motility control. Cell Commun Signal. 2010;8:23. https://doi.org/10.1186/1478-811X-8-23.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Davis RC, Maloney MT, Minamide LS, et al. Mapping cofilin-actin rods in stressed hippocampal slices and the role of cdc42 in amyloid-beta-induced rods. J Alzheimers Dis. 2009;18(1):35–50. https://doi.org/10.3233/JAD-2009-1122.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Fournier NM, Lee B, Banasr M, et al. Vascular endothelial growth factor regulates adult hippocampal cell proliferation through MEK/ERK- and PI3K/Akt-dependent signaling. Neuropharmacology. 2012;63(4):642–52. https://doi.org/10.1016/j.neuropharm.2012.04.033.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Tillo M, Erskine L, Cariboni A, et al. VEGF189 binds NRP1 and is sufficient for VEGF/NRP1-dependent neuronal patterning in the developing brain. Development. 2015;142(2):314–9. https://doi.org/10.1242/dev.115998.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Barber M, Andrews WD, Memi F, et al. Vascular-derived VEGFA promotes cortical interneuron migration and proximity to the vasculature in the developing forebrain. Cereb Cortex. 2018;28(7):2577–93. https://doi.org/10.1093/cercor/bhy082.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Cristy P. Brain-derived neurotrophic factor, depression, and physical activity: making the neuroplastic connection. Neural Plast. 2017;2017:1–17. https://doi.org/10.1155/2017/7260130.

    Article  CAS  Google Scholar 

  77. Jiao SS, Shen LL, Zhu C, et al. Brain-derived neurotrophic factor protects against tau-related neurodegeneration of Alzheimer’s disease. Transl Psychiat. 2016;6(10):e907. https://doi.org/10.1038/tp.2016.186.

    Article  CAS  Google Scholar 

  78. Bilkei-Gorzo A, Mauer D, Michel K, et al. Dynorphins regulate the strength of social memory. Neuropharmacology. 2014;77:406–13. https://doi.org/10.1016/j.neuropharm.2013.10.023.

    Article  CAS  PubMed  Google Scholar 

  79. Mikhail V, Juergen P, Christian W, et al. A functional polymorphism in the prodynorphin gene affects cognitive flexibility and brain activation during reversal learning. Front Behav Neurosci. 2015;9:172. https://doi.org/10.3389/fnbeh.2015.00172.

    Article  CAS  Google Scholar 

  80. Tejeda HA, Shippenberg TS, Henriksson R. The dynorphin/κ-opioid receptor system and its role in psychiatric disorders. Cell Mol Life Sci. 2016;69(6):857–96. https://doi.org/10.1007/s00018-011-0844-x.

    Article  CAS  Google Scholar 

  81. Stefanits H, Wesseling C, Kovacs GG. Loss of Calbindin immunoreactivity in the dentate gyrus distinguishes Alzheimer’s disease from other neurodegenerative dementias. Neurosci Lett. 2014;566:137–41. https://doi.org/10.1016/j.neulet.2014.02.026.

    Article  CAS  PubMed  Google Scholar 

  82. Verdaguer E, Brox S, Petrov D, et al. Vulnerability of calbindin, calretinin and parvalbumin in a transgenic/knock-in appswe/ps1de9 mouse model of alzheimer disease together with disruption of hippocampal neurogenesis. Exp Gerontol. 2015;69:176–88. https://doi.org/10.1016/j.exger.2015.06.013.

    Article  CAS  PubMed  Google Scholar 

  83. Szot P, Leverenz JB, Peskind ER, et al. Tyrosine hydroxylase and norepinephrine transporter mRNA expression in the locus coeruleus in Alzheimer's disease. Mol Brain Res. 2000;84(1):135–40. https://doi.org/10.1016/S0169-328X(00)00168-6.

    Article  CAS  PubMed  Google Scholar 

  84. Priyadarshini M, Kamal MA, Greig NH, et al. Alzheimer’s disease and type 2 diabetes: exploring the association to obesity and tyrosine hydroxylase. Cns Neurol Disord-Dr. 2012;11(4):482–9. https://doi.org/10.2174/187152712800792767.

    Article  CAS  Google Scholar 

  85. Cataldi M. The changing landscape of voltage-gated calcium channels in neurovascular disorders and in neurodegenerative diseases. Curr Neuropharmacol. 2013;11(3):276–97. https://doi.org/10.2174/1570159X11311030004.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Goodison WV, Frisardi V, Kehoe PG. Calcium channel blockers and Alzheimer’s disease: potential relevance in treatment strategies of metabolic syndrome. J Alzheimers Dis. 2012;30(Suppl 2):S2690–S282. https://doi.org/10.3233/JAD-2012-111664.

    Article  CAS  Google Scholar 

  87. Akiyama H, Tooyama I, Kawamata T, et al. Morphological diversities of CD44 positive astrocytes in the cerebral cortex of normal subjects and patients with Alzheimer's disease. Brain Res. 1993;632(1–2):249–59. https://doi.org/10.1016/0006-8993(93)91160-T.

    Article  CAS  PubMed  Google Scholar 

  88. Pinner E, Gruper Y, Ben ZM, et al. CD44 splice variants as potential players in Alzheimer's disease pathology. J Alzheimers Dis. 2017;58(4):1137–49. https://doi.org/10.3233/JAD-161245.

    Article  CAS  PubMed  Google Scholar 

  89. Uberti D, Cenini G, Bonini SA, et al. Increased CD44 gene expression in lymphocytes derived from Alzheimer disease patients. Neurodegener Dis. 2010;7(1–3):143–7. https://doi.org/10.1159/000289225.

    Article  CAS  PubMed  Google Scholar 

  90. Flashner E, Raviv U, Friedler A. The effect of tachykinin neuropeptides on amyloid β aggregation. Biochem Bioph Res Co. 2011;407(1):13–7. https://doi.org/10.1016/j.bbrc.2011.02.067.

    Article  CAS  Google Scholar 

  91. Wang N, Zhang Y, Xu L, et al. Relationship between Alzheimer’s disease and the immune system: a meta-analysis of differentially expressed genes. Front Neurosci. 2019;12:1026. https://doi.org/10.3389/fnins.2018.01026.

    Article  PubMed  PubMed Central  Google Scholar 

  92. Misrani A, Tabassum S, Long C. Oxytocin system in neuropsychiatric disorders: old concept, new insights. Sheng Li Xue Bao. 2017;69(2):196–206. https://doi.org/10.13294/j.aps.2016.0105.

    Article  PubMed  Google Scholar 

  93. Naja WJ, Aoun MP. Oxytocin and anxiety disorders: translational and therapeutic aspects. Curr Psychiat Rep. 2017;19(10):67. https://doi.org/10.1007/s11920-017-0819-1.

    Article  Google Scholar 

  94. Bowen MT, Neumann ID. Rebalancing the addicted brain: oxytocin interference with the neural substrates of addiction. Trends Neurosci. 2017;40(12):691–708. https://doi.org/10.1016/j.tins.2017.10.003.

    Article  CAS  PubMed  Google Scholar 

  95. Lin D, Pestova TV, Hellen CU, et al. Translational control by a small RNA: dendritic BC1 RNA targets the eukaryotic initiation factor 4A helicase mechanism. Mol Cell Biol. 2008;28(9):3008–19. https://doi.org/10.1128/MCB.01800-07.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Gene Expression Omnibus. https://www.ncbi.nlm.nih.gov/geo/. Accessed 12 Jun 2019.

  97. European Bioinformatics Institute’s (EBI) ArrayExpress-functional genomics database. https://www.ebi.ac.uk/arrayexpress/. Accessed 11 Jun 2019.

  98. Morpheus. https://www.software.broadinstitute.org/morpheus/. Accessed 6 Jun 2019.

  99. Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. Gene. 2000;25(1):25–9. https://doi.org/10.1038/75556.

    Article  CAS  Google Scholar 

  100. The Gene Ontology Consortium. The gene ontology resource: 20 years and still GOing strong. Nucleic Acids Res. 2019;47(D1):D330–8. https://doi.org/10.1093/nar/gky1055.

    Article  Google Scholar 

  101. Kanehisa M, Sato Y, Kawashima M, et al. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 2016;44(Database issue):D457–62. https://doi.org/10.1093/nar/gkv1070.

    Article  CAS  PubMed  Google Scholar 

  102. Kanehisa M, Furumichi M, Tanabe M, et al. KEGG: new perspectives on genomes, pathways, diseases and drugs. Nucleic Acids Res. 2017;45(D1):D353–61. https://doi.org/10.1093/nar/gkw1092.

    Article  CAS  PubMed  Google Scholar 

  103. Vastrik I, D'Eustachio P, Schmidt E, et al. Reactome: a knowledge base of biologic pathways and processes. Genome Biol. 2007;8(3):R39. https://doi.org/10.1186/gb-2007-8-3-r39.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Fabregat A, Sidiropoulos K, Viteri G, et al. Reactome pathway analysis: a high-performance in-memory approach. BMC Bioinformatics. 2017;18(1):142. https://doi.org/10.1186/s12859-017-1559-2.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Slenter DN, Kutmon M, Hanspers K, et al. WikiPathways: a multifaceted pathway database bridging metabolomics to other omics research. Nucleic Acids Res. 2018;46:D661–7. https://doi.org/10.1093/nar/gkx1064.

    Article  CAS  PubMed  Google Scholar 

  106. Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504. https://doi.org/10.1101/gr.1239303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Jüschke C, Dohnal I, Pichler P, et al. Transcriptome and proteome quantification of a tumor model provides novel insights into post-transcriptional gene regulation. Genome Biol. 2013;14(11):r133. https://doi.org/10.1186/gb-2013-14-11-r133.

    Article  PubMed  PubMed Central  Google Scholar 

  108. Tang B, Wang Q, Yang M, et al. Contigscape: a Cytoscape plugin facilitating microbial genome gap closing. BMC Genomics. 2013;14(1):289. https://doi.org/10.1186/1471-2164-14-289.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Su G, Morris JH, Demchak B, et al. Biological network exploration with Cytoscape 3. Curr Protoc Bioinformatics. 2014;47:8.13.1–8.13.24. https://doi.org/10.1002/0471250953.bi0813s47.

    Article  Google Scholar 

  110. Stringer KA, Mckay RT, Alla K, et al. Metabolomics and its application to acute lung diseases. Front Immuno. 2016;7:44. https://doi.org/10.3389/fimmu.2016.00044.

    Article  CAS  Google Scholar 

  111. Search Tool for the Retrieval of Interacting Genes database. https://www.string-db.org/. Accessed 13 Jun 2019.

  112. Szklarczyk D, Franceschini A, Wyder S, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43:D447–52. https://doi.org/10.1093/nar/gku1003.

    Article  CAS  PubMed  Google Scholar 

  113. Szklarczyk D, Morris JH, Cook H, et al. The STRING database in 2017: quality-controlled protein-protein association networks, made broadly accessible. Nucleic Acids Res. 2017;45:D362–8. https://doi.org/10.1093/nar/gkw937.

    Article  CAS  PubMed  Google Scholar 

  114. Kent WJ, Sugnet CW, Furey TS, et al. The human genome browser at UCSC. Genome Res. 2002;12(6):996–1006. https://doi.org/10.1101/gr.229102.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. University of Califorina Santa Cruz Genome Browser. https://genome.ucsc.edu/index.html. Accessed 17 Jun 2019.

  116. Berglund AC, Sjölund E, Ostlund G, et al. InParanoid 6: eukaryotic ortholog clusters with inparalogs. Nucleic Acids Res. 2008;36(Database issue):D263–6. https://doi.org/10.1093/nar/gkm1020.

    Article  CAS  PubMed  Google Scholar 

  117. The database of RNA-binding protein specificities. http://rbpdb.ccbr. utoronto.ca/index.php. Accessed 17 Jun 2019.

Download references

Acknowledgements

We are grateful to our colleagues for their contributions.

Funding

This work is supported by grants from the National Natural Science Foundation of China (31371082) and research fund from Harbin Institute of Technology at Weihai (HIT (WH).

Y200902).

Author information

Authors and Affiliations

Authors

Contributions

YT contributed to article writing and data analyses while DF helped with figure construction and manuscript proof, and ZY provided fund support and help with the technical problems. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Tingting Yan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

All the authors have consented for the publication.

Competing interests

The authors declare that they have no competing interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional files

Additional file 1

Table S1. Information for samples in the included datasets. (ArrayExpress datasets). Included detailed information of samples of hippocampus and entorhinal cortex regions extracted from GSE48350. (XLSX 14 kb)

Additional file 2

Table S2. Information for the DEGs identified from the GEO dataset (|log2FC| ≥ 1, p value< 0.05). Included detailed information of all DEGs screened from hippocampus and entorhinal cortex regions. (XLSX 31 kb)

Additional file 3

Table S3. Information for biological process analysis of DEGs from hippocampus. Included detailed information of results of biological process analysis of DEGs from hippocampus. (XLSX 32 kb)

Additional file 4

Table S4. Information for cellular component analysis of DEGs from hippocampus. Included detailed information of results of cellular component analysis of DEGs from hippocampus. (XLSX 21 kb)

Additional file 5

Table S5. Information for molecular function analysis of DEGs from hippocampus. Included detailed information of results of molecular function analysis of DEGs from hippocampus. (XLSX 17 kb)

Additional file 6

Table S6. Information for biological process analysis of DEGs from entorhinal cortex. Included detailed information of results of biological process analysis of DEGs from entorhinal cortex. (XLSX 16 kb)

Additional file 7

Table S7. Pathway enrichment analyses of DEGs. Included detailed information of pathway enrichment analyses of DEGs from hippocampus and entorhinal cortex. (XLSX 12 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yan, T., Ding, F. & Zhao, Y. Integrated identification of key genes and pathways in Alzheimer’s disease via comprehensive bioinformatical analyses. Hereditas 156, 25 (2019). https://doi.org/10.1186/s41065-019-0101-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41065-019-0101-0

Keywords