Identification of hub genes and molecular genetic mechanisms in Alzheimer’s Disease and Schizophrenia

DOI: https://doi.org/10.21203/rs.3.rs-1513208/v1

Abstract

Cognitive impairment and psychotic symptoms are both common in schizophrenia and Alzheimer's disease (AD), affecting more than half of the patients. Shared symptoms between AD and SCZ indicating similar cerebral pathophysiologies exist, common in both clinical and subclinical settings. Neurochemical alterations of the dopaminergic/cholinergic axis are important in schizophrenia and AD, as well as high-risk genes. Insights drawn from these observations enrich the understanding of the relationship between schizophrenia and AD, helping to further the symptomatic control of each disease.

1. Introduction

Alzheimer’s disease (AD) is the most frequently form of neurodegenerative disease, accounting for over half of all cases. Pateint may have deficts on memory, initiation, learning, and conceptualization[1]. People over 65-year-old are high incidence population in AD. And of all the cases, only 1% cases are early-onset familial mutations [2]. And due to the increased life expectancy, AD is expected to double every 20 years and reach 81 million cases in 2040[3]. As there has been remarkable progress with understanding of neurodegenerative disease mechanisms during recent years. It is elusive to search for true physiological mechanisms associated with formation of amyloid-β peptide (Aβ) plaques, as well as neurofibrillary tangles containing hyperphosphorylated tau [2]. It is recognized that early and even prodromal disease treatment timing is most effective intervention. However, this relies on biomarker-based diagnostics to help detect the presence of AD pathologies [4].

Schizophrenia (SCZ) is a mental disorder, with delusions, hallucinations, social withdrawal, and cognitive impairment as typical symptoms. SCZ patients would develope overall impaired functioning in not only self-care, but also work, school, parenting, independent living, and interpersonal relationships[5]. Incidence of SCZ is higher among relatives of patients, indicating the importance of genetic effects in disease.[6]. It has been reported that SCZ has multiple susceptibility genes act together with epigenetic and environmental factors, which have enabled large-scale studies to identify specific genetic links [7].

AD and SCZ often overlap in terms of the various clinical symptoms, from psychiatric symptoms to cognitive dysfunction[8], indicating a similar pathogenic mechanism exist between these two neurological disease [9]. And one significant step toward understanding the etiologies of the neuropsychiatric disorders within SCZ and AD is to identify reliable associations among the genetic variants. By then we can unlock their translational potential by understanding of their functional consequences and linking them to specific affected genes and biological processes.

2. Materials And Methods

2.1. Microarray Data of AD and SZ

Tow microarray profiles GSE5281 [10]and GSE53987[11] were obtained from Gene Expression Omnibus (GEO) database of National Center for Biotechnology Information (NCBI)[12]. GSE53987 data was based on GPL570 Platform (Affymetrix Human Genome U133 Plus 2.0 Array). Twenty-five samples from the hippocampus were used for further analysis, 15 SCZ and 10 normal samples included. As for data mining, GSE5281 is high reliable in quality, reasonable in experimental design, and can provide rich information. Data from GSE5281 was based on the GPL570 Platform (Affymetrix Human Genome U133 Plus 2.0 Array), including 161 chips covering 6 brain regions. In this study, 23 samples from hippocampus, containing 13 normal and 10 AD samples were chosen.

2.2. Data Preprocessing and Differential Expressed Analysis

GEO query (http://www.bioconductor.org/packages/release/bioc/html/GEOquery.html) was used to obtain the normalized expression profiles. Limma (Linear Models for Microarray Analysis) (https://bioconductor.org/packages/release/bioc/html/limma.html) packages were used to screen differentially expressed genes (DEGs). The Benjamini-Hochberg method was used to adjust the original p-values, and the false discovery rate (FDR) procedure was used to calculate the fold-changes (FC). Gene’s expression values of the |log2 FC|>1and adjusted p < 0.05 were used to filter the AD-DEGs. Meanwhile, the |log2 FC|>0.263 and the adjusted p < 0.05 were used to identify the SCZ-DEGs. Given that schizophrenia does not have gross brain pathology, the disorder involves subtle pathological changes within specific neural cell populations and in cell–cell communication [8]. Additionally, Venn diagramswe was calculated and made for co-DEGs for AD-DEGs and SZ-DEGs.

2.3. GO annotation and Pathway Enrichment Analysis

To assign gene ontology terms for their possible roles in the biological process, the Bioconductor R package ClusterProfiler [13] was applied to explore the functions among the genes of interest, with a cut-off criterion of an adjusted p-value (p < 0.05). The Kyoto Encyclopedia for Genes and Genomes (KEGG)-based screening (https://www.kegg.jp/) was also performed to identify the role of the DEGs and hub genes in various metabolic pathways. Gene Set Enrichment Analysis (GSEA) is used to assess whether a predefined gene set shows statistically significant differences between the two groups using an R package cluster profiler for AD and SCZ. Expression datasets that collapsed to the gene symbol and phenotype information were uploaded to the GSEA for enrichment analysis with default parameters. Enrichment results with an adjusted p value < 0.05 as well as an FDR < 0.25 were considered statistically significant.

2.4. PPI Network Construction and Hub Gene analysis

Protein-protein interactions (PPI) networks, including direct physical interaction of proteins and indirect functions, were predicted using the Search Tool for the Retrieval of Interacting Genes (STRING, https://string-db.org/) database. DEGs were mapped to STRING later, with the cut-off criterion of combined score greater than 0.4. The PPI network was visualized with the open-source bioinformatics software Cytoscape (version 3.7.2). CytoHubba, a Cytoscape plugin, was used to obtain the network center nodes (hub genes). The proteins expressed by the central node were usually an important protein/gene with critical physiological function. Ten genes in the maximum correlation criterion (MCC) were chosen by the CytoHubba plugin and sequentially ordered. A redder color represents more forward rankings.

3. Results

3.1 Identification of DEGs in AD and SZ

After normalization, the mean gene expression values for each sample in AD and SZ were fundamental equal. By limma package (Version 3.26.9),2281 DEGs, including 601 upregulated and 1680 downregulated DEGs, were identified in GSE58793 (SZ)(Figure.1.a). While in GSE5281 (AD) 2769 DEGs, including 1437 upregulated and 1332 downregulated DEGs, were identified(Figure.1.b). Then we mapped these top 50 differentially expressed genes into heatmaps to assess the differences in expression between disease group and control group (Figure.1.c). As shown in (Figure.1.d), 613 overlapping of DEGs were found in SZ and AD, including 222 co-upregulated DEGs and 391 co-downregulated DEGs.

3.2. Functional annotation of DEGs

Then we explore the potentially altered functional characteristics associated with the DEGs in AD and SCZ, and Gene Ontology (GO) analysis was carried out for the differences in the biological processes between the disease group and the control group in AD and SCZ respectively.

In AD, genes upregulated in the disease group were major involved in protein modification, including regulation of chromatin organization, positive regulation of histone deacetylation, regulation of histone modification, and positive regulation of protein deacetylation. In contrast, downregulated genes in the AD disease group were tiedly related to metabolic process, including ATP metabolic process, cellular respiration, RNA catabolic process and regulation of mRNA metabolic/cellular amino acid metabolic process. Moreover, most of the positively related genes were enriched in KEGG terms including long-term depression, long-term potentiation. And the negatively correlated genes were enriched within the citrate cycle (TCA cycle), biosynthesis of amino acids, and amyotrophic lateral sclerosis. Next, validation was carried out by GSEA analyses, showing highly expressed genes that were significantly associated with long-term depression and estrogen signaling pathway, long-term potentiation, dopaminergic synapse, and Amoebiasis. The low expression gene group was significantly associated with oxidative phosphorylation, Huntington disease, proteasome, Parkinson disease, carbon metabolism, metabolic pathways, and Amyotrophic lateral sclerosis.

In SCZ, genes upregulated and downregulated in the GO terms and the KEGG pathways are shown as Fig. 3. Our GSEA analysis results indicated that the low expression genes were distinctly enriched in pathways of neurodegeneration disease, including AD, prion disease, Huntington disease(HD), Amyotrophic lateral sclerosis(ALS).

Further, we conducted the KEGG pathway and GO enrichment analysis of the 222 co-upregulated and 391 co-downregulated DEGs to study the functions of the 613 overlapping genes. And 222 overlapping co-upregulated genes within the biological processes (BP) were found closely related to the regulation of chromatin organization, positive regulation of histone deacetylation, regulation of histone modification, positive regulation of protein deacetylation, and regulation of protein-containing complex assembly. (Table 1) (Figure.2a). The BP of the 391 co-downregulated DEGs was primarily related to the regulation and activation of the innate immune response, ATP metabolic process, anaphase-promoting complex-dependent catabolic process, antigen processing and presentation of exogenous peptide antigen and regulation of stem cell differentiation. (Table 2) (Figure.2b). Moreover, in the KEGG pathway enrichment analysis, positively related co-DEGs were enriched within the MAPK signaling,cancer and the mTOR signaling pathways (Figure.2.c), while the negatively correlated genes were enriched within pathways of multiple neurodegeneration disease, AD,HD, ALS included(Table 3) (Figure.2d).

3.3 PPI Network Construction and Hub Genes Identification

Overlapping genes were analyzed by PPI network, 222 co-upregulated DEGs and 391 co-downregulated DEGs were established using the STRING database. In PPI networks, hub genes were defined as genes with stronger interactions with numerous other genes. And hub genes are potential drivers of the pathology of the diseases. In order to screen hub genes among all the DEGs via the MCC scores, cytoHubba plugin for Cytoscape was used. Interestingly, all these 10 hub genes got from screening were downregulated co-DEGs (Figure.3). And proteasome subunit alpha 5 (PSMA5), proteasome subunit beta 7 (PSMB7), proteasome 26S subunit, non-ATPase 12 (PSMD12), proteasome subunit alpha 1 (PSMA1), proteasome 26S subunit, ATPase 3 (PSMC3), proteasome 26S subunit, non-ATPase 4 (PSMD4), proteasome subunit beta 3 (PSMB3), proteasome subunit beta 1 (PSMB1), proteasome 26S subunit, non-ATPase 1 (PSMD1), and proteasome subunit alpha 7 (PSMA7) are the top 10 genes with the highest MCC sores, respectively .

3.4 GO Enrichment Analysis of the hub genes

All ten hub genes are downregulated DEGs both in SCZ and AD. Using the R package clusterprofiler. And among those genes, the top 10 GO terms were primarily related to the regulation of hematopoietic stem cell differentiation, regulation of cellular amino acid metabolic process, TAP-dependent,antigen processing and presentation of exogenous peptide antigen via MHC class I, antigen processing and presentation of exogenous peptide antigen via MHC class I, regulation of cellular amine metabolic process, regulation of transcription from RNA polymerase II promoter in response to hypoxia, anaphase-promoting complex-dependent catabolic process, regulation of hematopoietic progenitor cell or stem cell differentiation, and SCF-dependent proteasomal ubiquitin-dependent protein catabolic process. (Figure.4) (Table 4).

4. Discussion

This study foucsed on investigation of the expression association files between AD and SCZ. The interrelated mechanisms of AD and SZ are mainly focused on defective mitochondrial metabolism [14, 15]. The co-expression patterns of these different pathways of the each diseases are rarely studied. Therefore, specific genes and pathways especially in the histopathological highly related hippocampal regions of these neurological diseases are urgently need to be identified.

4.1 Biological function of the 613 Overlapping Genes

Some well-known pathways associated with AD and SCHIZ, such as the ATP metabolic and cellular amino acid metabolic processes, have also been observed in our study. These processes will not be discussed in this article [16, 17]. GO enrichment analysis of the co-DEGs also involved positive regulation in chromatin organization, histone deacetylation, and protein deacetylation, which, to some extent, related to epigenetics modifications. Modifications of histone proteins are associated with chromatin structure and play a pivotal role in epigenetic regulation of transcription[18]. Histone acetylation disrupts the structured arrangement of histone proteins and loosens the chromatin structure. This process makes it more accessible to transcription binding, while histone deacetylation removes the acetyl groups and generally links to chromatin inactivation. Experimental evidence suggests that treatment with histone deacetylase inhibitors ameliorates neuronal deficiencies and enhances synaptic plasticity, memory, and learning as a promising new strategy for neurodegenerative and psychiatric disorders[19]. Protein-protein interaction, deacetylation mechanism of non-histone protein, histone post-translational modifications and direct association with disease proteins have also been proved to link to neuronal imbalance, as well as transcriptional regulation[20]. These results are consistent with our analysis, and provides an open mechanism research point in epigenetic modifications of neuronal disease development. Therefore, new insights into the etiology of the pathogenesis of AD and SCHIZ, as well as new avenues for developing therapeutic strategies, remain to be explored[21].

In KEGG enrichment, the upregulated co-DEGs were enriched within the interleukin-17 (IL-17) signaling pathway (Table.6). Interleukin-17a (IL-17a) was produced by Th17 cells. For pregnant mice suffering immune system activation due to infections or autoinflammatory syndromes, IL-17a may increase the risk for neurodevelopmental disorders in offspring[22]. At the interface of neurological and psychiatric disorders, immune processes play an important role in central nervous system health and disease; the immune system contributes to overall CNS environmental stability, brain reserves and resilience[23]. Genome-wide association studies (GWAS) in patients with neurodegenerative disease are preferentially enriched within the enhancer sequences implicating innate immune processes [2428]. Preclinical, experimental, and bioinformatic analysis showed that activation of the immune system is accompanied by AD pathology, which contributes to the pathogenesis of AD[29]. Over the past three decades, the causal relationship between pathogens in Alzheimer's disease has been repeatedly postulated[30, 31]. Associations between Alzheimer's disease and infectious burden was investigated in a A cross-sectional study. Results from this study showed that patients with Alzheimer’s disease had higher prevalence in prior infection with CMV, HSV-1, B. burgdorferi, C. pneumoniae, and H. pylori than age-matched healthy controls[25, 32]. Pathological assumption was then raised that pathogens may cause neurological damage by eliciting neuroinflammation in CNS by directly cross the blood-brain barrier[30]. Moreover, elders with a higher IB would develope worse cognition and higher serum Aβ levels, not only AD patients but also healthy controls[32]. As a result, more than one possibility can be proposed for the association between suspected pathogens and Alzheimer's disease. One is that Alzheimer's patients are more susceptible to microbial infections. Bacterial lipopolysaccharide and viral surface proteins shared same receptors that can sense pathogen-associated molecular patterns can both be triggered by Aβ aggregates [33]. The other is that microbial infections have a contributory role in progression of Alzheimer’s disease[34]. A genome-wide association study study examined the association between schizophrenia and markers near the region of the major histocompatibility complex (MHC) on chromosome 6. Many immune-related genes was involved in this association study, including antigen presentation and inflammatory mediators[35].

4.2. Hub Genes from Gene Co Expression Network

Among the co-DEGs, 10 hub genes were selected according to the maximum correlation criterion (MCC) scores (Table 2). Here, we focus on the analysis of the following genes: PSMA5, PSMA7, PSMA1, PSMD4, PSMD1, PSMD12, PSMB7, PSMC3, PSMB1, and PSMB3

PSMA5, PSMA7, PSMA1, PSMB7, PSMB1, PSMB3, and PSMC3 are the subunit of the 20S proteasome, while PSMD4, PSMD1, and PSMD12 are the subunits of 26S proteasome. Apart from mitochondria, other important aging-related transcriptomic changes were the downregulation of genes related to proteasomal functions [36]. The proteasome is responsible for a large number of protein turnovers, especially degradation of oxidized proteins and short life, which mediates the degradation of proteins. The alpha-20S subunits and beta-20S proteasome subunits are responsible for regulating proteasome activity and mediating the different proteolytic specificities of the proteasome respectively. There is evidence that proteasome inhibition contributes to increased oxidative damage, elevated intracellular levels of protein oxidation, and the induction of oxidative stress. Therefor psychiatric disorders and neurodegenerative diseases both had oxidative damage and neuronal loss[3740]. Reactive oxygen species (ROS) cause nucleic acid breakage, lipid peroxidation, polysaccharide depolymerization, enzyme inactivation,and a host of other destructive processes [41, 42]. Furthermore, elevated ROS concentrations hinders mitochondrial activities, thus triggering the generation of Aβ [43, 44]. Yili Wu et al. used western blots and quantitative PCR to validate that PSMA5 and PSMB7 were downregulated via the over-expression of amyloid precursor proteins in HEK 293 cells [45]. Improving ubiquitin proteasome system activities recovers proteasome activities and ameliorates cell survival of HD-patient derived neurons, which is also a neurodegenerative disease[46]. These facts are consistent with our results of the 10 hub genes involved in the AD and SCZ.

In summary, we have provided information on the DEGs and hub genes involved in both AD and SCZ by using bioinformatics. AD patients have a more remarkable hippocampus gene expression than SCZ patients. Traditional boundaries between neurological and psychiatric disorders become blurred because of the common emerged pathways. However, the limitation is that expression of these genes in the different AD and SCZ subtypes were not involved in this analysis. Another limitation is that, we obtained the criteria of |FC|>1.2 in reducing the number of false negative DEGs in SCZ. And consequently, we may involve many insignificance genes in this study. While our work showed the usefulness and benefit of microarray analysis in extracting DEGs and hub genes for possible new targets of AD and SCZ. It is imperative to improve experimental analysis and prospective clinical studies. And the new insight of the underlying molecular mechanisms shared by AD and SCZ can guild subsequent experimental studies.

Declarations

Data Availability Statement

The datasets generated during and/or analysed during the current study are available in the GEO, KEGG and STRING repository. [ http://www.bioconductor.org/packages/release/bioc/html/GEOquery.html, https://www.kegg.jp/, https://string-db.org/].

Acknowledgments

GEO, KEGG and STRING belong to public databases. We acknowledge all these four database for providing their platforms and contributors for uploading their meaningful datasets.Our study is based on open source data, so there are no ethical issues and other conflicts of interest. This research was supported by grants from the Science, Technology, and Innovation Commission of Shenzhen Municipality Technology Fund (Nos.JCYJ20170818093322718). Shenzhen second people's hospital clinical research project (20203357027).The authors also thank AiMi Academic Services (www.aimieditor.com) for the English language editing and review services.

Disclosure

GEO, KEGG, and STRING belong to public databases. We acknowledge all these four database for providing their platforms and contributors for uploading their meaningful datasets.Our study is based on open source data, so there are no ethical issues and other.

GEO (Gene Expression Omnibus):https://www.ncbi.nlm.nih.gov/geo/. GEO is a public functional genomics data repository supporting MIAME-compliant data submissions. 

KEGG (Kyoto Encyclopedia of Genes and Genomesis): https://www.kegg.jp/. KEGG is a bioinformatics resource for linking genomes to life and the environment.

STRING (Search Tool for the Retrieval of Interacting Genes): https://string-db.org/. STRING is a protein-protein interaction networks searching database, provide a rich source of drug targets for the development of new generation of clinical therapeutic.

Consent for publication

The author agrees to publication of this article in Journal of Molecular Neuroscience. The work has not been published before. And its publication has been approved by all co-authors.

Competing interests

The authors declare that there is no conflict of interest.

Funding

This research was supported by grants from the Science, Technology, and Innovation Commission of Shenzhen Municipality Technology Fund (Nos.JCYJ20170818093322718). Shenzhen second people's hospital clinical research project (20203357027).

Authors' contributions

Under supervision by Lijie Ren and Ting Chen; Yixuan Zeng performed sample preparation and data analysis; Ting Chen performed sample preparation. Yuxi Zheng performed calculations and figure preparation. Yixuan Zeng and Yuxi Zheng performed the manuscript writing. All authors read and contributed to the manuscript. All authors approved the final manuscript.

References

  1. Masliah E: Mechanisms of synaptic dysfunction in Alzheimer's disease. Histol Histopathol 1995, 10(2):509-519.
  2. Blennow K, de Leon MJ, Zetterberg H: Alzheimer's disease. Lancet 2006, 368(9533):387-403.
  3. Ferri CP, Prince M, Brayne C, Brodaty H, Fratiglioni L, Ganguli M, Hall K, Hasegawa K, Hendrie H, Huang Y et al: Global prevalence of dementia: a Delphi consensus study. Lancet 2005, 366(9503):2112-2117.
  4. Desai AK, Grossberg GT: Diagnosis and treatment of Alzheimer's disease. Neurology 2005, 64(12 Suppl 3):S34-39.
  5. Mueser KT, McGurk SR: Schizophrenia. Lancet 2004, 363(9426):2063-2072.
  6. McGuffin P, Owen MJ, Farmer AE: Genetic basis of schizophrenia. Lancet 1995, 346(8976):678-682.
  7. Bayer TA, Falkai P, Maier W: Genetic and non-genetic vulnerability factors in schizophrenia: the basis of the "two hit hypothesis". Journal of psychiatric research 1999, 33(6):543-548.
  8. Kahn RS, Sommer IE, Murray RM, Meyer-Lindenberg A, Weinberger DR, Cannon TD, O'Donovan M, Correll CU, Kane JM, van Os J et al: Schizophrenia. Nat Rev Dis Primers 2015, 1:15067.
  9. Takamatsu Y, Ho G, Waragai M, Wada R, Sugama S, Takenouchi T, Masliah E, Hashimoto M: Transgenerational Interaction of Alzheimer's Disease with Schizophrenia through Amyloid Evolvability. Journal of Alzheimer's disease : JAD 2019, 68(2):473-481.
  10. Liang WS, Dunckley T, Beach TG, Grover A, Mastroeni D, Walker DG, Caselli RJ, Kukull WA, McKeel D, Morris JC et al: Gene expression profiles in anatomically and functionally distinct regions of the normal aged human brain. Physiol Genomics 2007, 28(3):311-322.
  11. Hu X, Zhang B, Liu W, Paciga S, He W, Lanz TA, Kleiman R, Dougherty B, Hall SK, McIntosh AM et al: A survey of rare coding variants in candidate genes in schizophrenia by deep sequencing. Molecular psychiatry 2014, 19(8):857-858.
  12. Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, Kim IF, Soboleva A, Tomashevsky M, Edgar R: NCBI GEO: mining tens of millions of expression profiles--database and tools update. Nucleic acids research 2007, 35(Database issue):D760-765.
  13. Yu G, Wang LG, Han Y, He QY: clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS 2012, 16(5):284-287.
  14. Holper L, Ben-Shachar D, Mann JJ: Multivariate meta-analyses of mitochondrial complex I and IV in major depressive disorder, bipolar disorder, schizophrenia, Alzheimer disease, and Parkinson disease. Neuropsychopharmacology : official publication of the American College of Neuropsychopharmacology 2019, 44(5):837-849.
  15. Nandi A, Yan LJ, Jana CK, Das N: Role of Catalase in Oxidative Stress- and Age-Associated Degenerative Diseases. Oxid Med Cell Longev 2019, 2019:9613090.
  16. Butterfield DA, Halliwell B: Oxidative stress, dysfunctional glucose metabolism and Alzheimer disease. Nat Rev Neurosci 2019, 20(3):148-160.
  17. Griffin JW, Bradshaw PC: Amino Acid Catabolism in Alzheimer's Disease Brain: Friend or Foe? Oxid Med Cell Longev 2017, 2017:5472792.
  18. Konsoula Z, Barile FA: Epigenetic histone acetylation and deacetylation mechanisms in experimental models of neurodegenerative disorders. J Pharmacol Toxicol Methods 2012, 66(3):215-220.
  19. Abel T, Zukin RS: Epigenetic targets of HDAC inhibition in neurodegenerative and psychiatric disorders. Current opinion in pharmacology 2008, 8(1):57-64.
  20. Gupta R, Ambasta RK, Kumar P: Pharmacological intervention of histone deacetylase enzymes in the neurodegenerative disorders. Life sciences 2020, 243:117278.
  21. Harrison IF, Dexter DT: Epigenetic targeting of histone deacetylase: therapeutic potential in Parkinson's disease? Pharmacology & therapeutics 2013, 140(1):34-52.
  22. Kim S, Kim H, Yim YS, Ha S, Atarashi K, Tan TG, Longman RS, Honda K, Littman DR, Choi GB et al: Maternal gut bacteria promote neurodevelopmental abnormalities in mouse offspring. Nature 2017, 549(7673):528-532.
  23. Pape K, Tamouza R, Leboyer M, Zipp F: Immunoneuropsychiatry - novel perspectives on brain disorders. Nat Rev Neurol 2019, 15(6):317-328.
  24. Shi J, Levinson DF, Duan J, Sanders AR, Zheng Y, Pe'er I, Dudbridge F, Holmans PA, Whittemore AS, Mowry BJ et al: Common variants on chromosome 6p22.1 are associated with schizophrenia. Nature 2009, 460(7256):753-757.
  25. Lampron A, Elali A, Rivest S: Innate immunity in the CNS: redefining the relationship between the CNS and Its environment. Neuron 2013, 78(2):214-232.
  26. Castro-Gomez S, Binder J, Heneka MT: [Neuroinflammation as motor of Alzheimer's disease]. Der Nervenarzt 2019, 90(9):898-906.
  27. Griciuc A, Tanzi RE: The role of innate immune genes in Alzheimer's disease. Current opinion in neurology 2021.
  28. Meyer-Luehmann M, Prinz M: Myeloid cells in Alzheimer's disease: culprits, victims or innocent bystanders? Trends Neurosci 2015, 38(10):659-668.
  29. Zhang B, Gaiteri C, Bodea LG, Wang Z, McElwee J, Podtelezhnikov AA, Zhang C, Xie T, Tran L, Dobrin R et al: Integrated systems approach identifies genetic nodes and networks in late-onset Alzheimer's disease. Cell 2013, 153(3):707-720.
  30. Panza F, Lozupone M, Solfrizzi V, Watling M, Imbimbo BP: Time to test antibacterial therapy in Alzheimer's disease. Brain : a journal of neurology 2019, 142(10):2905-2929.
  31. Itzhaki RF, Lathe R, Balin BJ, Ball MJ, Bearer EL, Braak H, Bullido MJ, Carter C, Clerici M, Cosby SL et al: Microbes and Alzheimer's Disease. Journal of Alzheimer's disease : JAD 2016, 51(4):979-984.
  32. Bu XL, Yao XQ, Jiao SS, Zeng F, Liu YH, Xiang Y, Liang CR, Wang QH, Wang X, Cao HY et al: A study on the association between infectious burden and Alzheimer's disease. European journal of neurology 2015, 22(12):1519-1525.
  33. Heneka MT, Golenbock DT, Latz E: Innate immunity in Alzheimer's disease. Nat Immunol 2015, 16(3):229-236.
  34. Walker KA, Gottesman RF, Wu A, Knopman DS, Gross AL, Mosley TH, Jr., Selvin E, Windham BG: Systemic inflammation during midlife and cognitive change over 20 years: The ARIC Study. Neurology 2019, 92(11):e1256-e1267.
  35. Schizophrenia Working Group of the Psychiatric Genomics C: Biological insights from 108 schizophrenia-associated genetic loci. Nature 2014, 511(7510):421-427.
  36. Cho BA, Yoo SK, Song YS, Kim SJ, Lee KE, Shong M, Park YJ, Seo JS: Transcriptome Network Analysis Reveals Aging-Related Mitochondrial and Proteasomal Dysfunction and Immune Activation in Human Thyroid. Thyroid 2018, 28(5):656-666.
  37. Jiang J, Peng C, Sun L, Li J, Qing Y, Hu X, Yang X, Li Y, Xu C, Zhang J et al: Leukocyte Proteomic Profiling in First-Episode Schizophrenia Patients: Does Oxidative Stress Play Central Roles in the Pathophysiology Network of Schizophrenia? Antioxid Redox Signal 2019, 31(8):579-588.
  38. Ding Q, Dimayuga E, Keller JN: Proteasome regulation of oxidative stress in aging and age-related diseases of the CNS. Antioxid Redox Signal 2006, 8(1-2):163-172.
  39. Ding Q, Keller JN: Proteasomes and proteasome inhibition in the central nervous system. Free Radic Biol Med 2001, 31(5):574-584.
  40. Pratico D: Oxidative stress hypothesis in Alzheimer's disease: a reappraisal. Trends Pharmacol Sci 2008, 29(12):609-615.
  41. Yan MH, Wang X, Zhu X: Mitochondrial defects and oxidative stress in Alzheimer disease and Parkinson disease. Free Radic Biol Med 2013, 62:90-101.
  42. Nagano T, Mizuno M, Morita K, Nawa H: Pathological Implications of Oxidative Stress in Patients and Animal Models with Schizophrenia: The Role of Epidermal Growth Factor Receptor Signaling. Curr Top Behav Neurosci 2016, 29:429-446.
  43. Heiss WD, Szelies B, Kessler J, Herholz K: Abnormalities of energy metabolism in Alzheimer's disease studied with PET. Ann N Y Acad Sci 1991, 640:65-71.
  44. Xiao SF, Cao QY, Xue HB, Liu YC, Zuo CT, Jiang KD, Zhang MY: [Measurement of regional cerebral metabolism rate of glucose in patients with Alzheimer's disease in different levels of severity]. Zhonghua Yi Xue Za Zhi 2005, 85(42):2975-2979.
  45. Wu Y, Zhang S, Xu Q, Zou H, Zhou W, Cai F, Li T, Song W: Regulation of global gene expression and cell proliferation by APP. Sci Rep 2016, 6:22460.
  46. Seo H, Sonntag KC, Kim W, Cattaneo E, Isacson O: Proteasome activator enhances survival of Huntington's disease neuronal model cells. PLoS One 2007, 2(2):e238.

Tables

Table.1 The Gene Ontology (GO) terms enrichment among co-upregulated genes in the biological process (BP).

ID

Description    

GeneRatio

pvalue

p.adjust

FDR

geneID

GO:0090311

regulation of protein deacetylation

7/198

4.08E-07

0.001217298

0.001089656

2033/25836/4137/6497/7422/26993/604

GO:1902275

regulation of chromatin organization

11/198

7.00E-06

0.010439652

0.00934498

55904/4524/4297/25836/6497/55729/201163/7
422/26993/4300/604

GO:0031063

regulation of histone deacetylation

5/198

1.17E-05

0.011638473

0.010418097

25836/6497/7422/26993/604

GO:0033044

regulation of chromosome organization

14/198

2.51E-05

0.014084027

0.012607217

55904/4524/4297/25836/4137/6497/7756/
4926/55729/201163/7422/26993/4300/604

GO:0031065

positive regulation of histone deacetylation

4/198

2.52E-05

0.014084027

0.012607217

25836/7422/26993/604

GO:0031056

regulation of histone modification

9/198

2.83E-05

0.014084027

0.012607217

55904/4524/4297/25836/6497/201163/
7422/26993/604

GO:0090312

positive regulation of protein deacetylation

4/198

6.12E-05

0.026080652

0.023345911

25836/7422/26993/604

GO:0006476

protein deacetylation

7/198

7.30E-05

0.027197375

0.024345538

2033/25836/4137/6497/7422/26993/604

GO:0043254

regulation of protein-containing complex assembly

15/198

8.31E-05

0.027530434

0.024643673

4131/6249/2033/4137/6709/57600/9113/
2017/4926/55729/51466/8780/7422/253260/5894

GO:1905269

positive regulation of chromatin organization

7/198

1.28E-04

0.036764115

0.032909138

55904/4297/25836/55729/7422/26993/604

GO:0035601

protein deacylation

7/198

1.36E-04

0.036764115

0.032909138

2033/25836/4137/6497/7422/26993/604


Table.2 The gene ontology(GO terms) enrichment among co-upregulated genes in the biological process (BP)

ID

Description    

GeneRatio

pvalue

p.adjust

FDR

geneID

GO:0046034

ATP metabolic process

31/376

1.76E-13

3.21E-10

2.77E-10

51422/30968/2597/29796/5213/318/523/539/2821/
4729/2026/506/4723/1738/10762/7385/8604/4706/
509/6622/2023/126328/4722/5230/10059/7384/6472/
526/4716/498/518

GO:0002478

antigen processing and presentation of exogenous peptide antigen

23/376

1.06E-12

9.66E-10

8.35E-10

5689/5691/10540/3831/5707/5719/5682/5695/5710/
1778/5702/1173/22920/2923/5693/10121/5694/1780/
5686/8905/5718/5688/9861

GO:0019884

antigen processing and presentation of exogenous antigen

23/376

2.71E-12

1.98E-09

1.71E-09

5689/5691/10540/3831/5707/5719/5682/5695/5710/
1778/5702/1173/22920/2923/5693/10121/5694/1780/
5686/8905/5718/5688/9861

GO:0031145

anaphase-promoting complex-dependent catabolic process

16/376

6.92E-12

3.83E-09

3.31E-09

5689/5691/5707/8452/5719/5682/5695/5710/5702/
996/5693/5694/5686/5718/5688/9861

GO:0048002

antigen processing and presentation of peptide antigen

23/376

7.35E-12

3.83E-09

3.31E-09

5689/5691/10540/3831/5707/5719/5682/5695/
5710/1778/5702/1173/22920/2923/5693/10121/
5694/1780/5686/8905/5718/5688/9861

GO:0060218

hematopoietic stem cell differentiation

16/376

1.48E-11

6.73E-09

5.81E-09

5689/5691/5707/5591/5719/5682/5695/5710/5702/
5693/5694/4005/5686/5718/5688/9861

GO:0002479

antigen processing and presentation of exogenous peptide antigen via MHC class I, TAP-dependent

15/376

1.81E-11

6.90E-09

5.96E-09

5689/5691/5707/5719/5682/5695/5710/5702/
2923/5693/5694/5686/5718/5688/9861

GO:0006521

regulation of cellular amino acid metabolic process

14/376

1.89E-11

6.90E-09

5.96E-09

5689/5691/5707/5719/5682/5695/5710/5702/
5693/5694/5686/5718/5688/9861

GO:2000736

regulation of stem cell differentiation

18/376

2.09E-11

6.94E-09

6.00E-09

5689/5691/5707/11137/5591/5719/5682/
5695/5710/11051/5702/5693/5694/4005/5686/
5718/5688/9861

GO:0042590

antigen processing and presentation of exogenous peptide antigen via MHC class I

15/376

4.82E-11

1.46E-08

1.27E-08

5689/5691/5707/5719/5682/5695/5710/5702/
2923/5693/5694/5686/5718/5688/9861

GO:0002218

activation of innate immune response

19/376

7.80E-11

2.19E-08

1.89E-08

5689/5691/5707/5591/5719/5682/6421/
5695/10084/5710/5702/5693/5694/6885/
5686/4841/5718/5688/9861


Table.3 The KEGG pathway enrichment analysis of the negatively correlated genes

ID

Description    

GeneRatio

pvalue

p.adjust

FDR

geneID

hsa05012

Parkinson disease

44/299

9.913677e-19

2.726261e-16

2.462766e-16

4137/3800/203068/5689/5691/3831/
29796/5707/7345/5719/10381/5682/
7416/539/5695/4729/5710/291/506/
4723/5601/7385/4706/509/6622/5702/
126328/4722/7384/7417/5693/5694/
6390/4716/498/5686/7419/292/801/518/
5718/5688/9861/7277

hsa05016

Huntington disease

47/299

2.078077e-17

2.857355e-15

2.581190e-15

2033/3800/203068/5689/5691/10540/
3831/29796/5707/5719/10381/5682/
7416/539/5695/4729/5710/291/506/
4723/6647/5601/7385/29982/4706/
509/5702/1173/126328/4722/7384/7417/
5693/10121/5694/6390/4716/498/5686/
5434/7419/292/518/5718/5688/9861/7277

hsa05020

Prion disease

41/299

6.418551e-15

5.461833e-13

4.933943e-13

3800/203068/5689/5691/3831/29796/
5707/5719/10381/5682/7416/539/5695/
4729/5710/291/506/4723/6647/5601/7385/
4706/509/5702/126328/4722/7384/7417/
5693/5694/6390/4716/498/5686/7419/292/
518/5718/5688/9861/7277

hsa05010

Alzheimer disease

48/299

7.944484e-15

5.461833e-13

4.933943e-13

4137/3800/208/5894/10313/203068/5689/
5691/3831/2597/29796/5707/5719/10381/
5682/7416/539/5695/4729/5710/291/506/
4723/5601/7385/29982/4706/509/6622/
5702/126328/4722/7384/7417/5693/5694/
6390/4716/498/5686/7419/292/801/518/
5718/5688/9861/7277

hsa05014

Amyotrophic lateral sclerosis

45/299

3.951487e-13

2.173318e-11

1.963265e-11

3800/9782/56893/203068/5689/5691/
10540/3831/10189/29796/5707/5719/
10381/5682/7416/539/5695/4729/5710/
506/4723/6647/10762/7385/5861/29982/
4706/509/5702/126328/4722/7384/5693/
10121/5694/6390/4716/498/5686/518/5718/
5688/6428/9861/7277

hsa05022

Pathways of neurodegeneration - multiple diseases

50/299

8.360702e-12

3.831988e-10

3.461624e-10

4137/3800/203068/5689/5691/3831/29796/
5707/7345/5719/10381/5682/7416/539/5695/
4729/5710/291/506/4723/5601/7385/4706/
509/6622/5702/126328/4722/7384/7417/5693/
5694/6390/4716/498/5686/7419/292/801/518/
5718/5688/9861/7277

hsa05017

Spinocerebellar ataxia

25/299

5.924485e-11

2.327476e-09

2.102524e-09

2033/3800/203068/5689/5691/10540/3831/
29796/5707/5719/10381/5682/7416/539/5695/
4729/5710/291/506/4723/6647/5601/7385/29982/
4706/509/5702/1173/126328/4722/7384/7417/
5693/10121/5694/6390/4716/498/5686/5434/
7419/292/518/5718/5688/9861/7277

hsa03050

Proteasome

14/299

5.381144e-10

2.726261e-16

1.670987e-08

3800/203068/5689/5691/3831/29796/5707/
5719/10381/5682/7416/539/5695/4729/5710/
291/506/4723/6647/5601/7385/4706/509/5702/
126328/4722/7384/7417/5693/5694/6390/4716/
498/5686/7419/292/518/5718/5688/9861/7277

hsa00190

Oxidative phosphorylation

20/299

7.427384e-08

2.857355e-15

2.050132e-06

4137/3800/208/5894/10313/203068/5689/
5691/3831/2597/29796/5707/5719/10381/
5682/7416/539/5695/4729/5710/291/506/4723/
5601/7385/29982/4706/509/6622/5702/126328/
4722/7384/7417/5693/5694/6390/4716/498/
5686/7419/292/801/518/5718/5688/9861/7277

hsa01200

Carbon metabolism

18/299

2.719592e-07

5.461833e-13

6.756038e-06

3800/9782/56893/203068/5689/5691/10540/
3831/10189/29796/5707/5719/10381/5682/7416/
539/5695/4729/5710/506/4723/6647/10762/
7385/5861/29982/4706/509/5702/126328/4722/
7384/5693/10121/5694/6390/4716/498/5686/518/
5718/5688/6428/9861/7277


Table.4 GO Enrichment Analysis of the hub genes

ID

Description    

GeneRatio

pvalue

p.adjust

FDR

geneID

GO:0006521

regulation of cellular amino acid metabolic process

10/10

8.14E-26

1.12E-23

1.88E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:1902036

regulation of hematopoietic stem cell differentiation

10/10

3.96E-25

2.41E-23

4.04E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0002479

antigen processing and presentation of exogenous peptide antigen via MHC class I, TAP-dependent

10/10

5.28E-25

2.41E-23

4.04E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0061418

regulation of transcription from RNA polymerase II promoter in response to hypoxia

10/10

6.99E-25

2.41E-23

4.04E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0042590

antigen processing and presentation of exogenous peptide antigen via MHC class I

10/10

1.05E-24

2.75E-23

4.62E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0033238

regulation of cellular amine metabolic process

10/10

1.20E-24

2.75E-23

4.62E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0031145

anaphase-promoting complex-dependent catabolic process

10/10

1.55E-24

3.05E-23

5.12E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:1901532

regulation of hematopoietic progenitor cell differentiation

10/10

2.25E-24

3.89E-23

6.53E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0060218

hematopoietic stem cell differentiation

10/10

2.55E-24

3.91E-23

6.56E-24

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702

GO:0031146

SCF-dependent proteasomal ubiquitin-dependent protein catabolic process

10/10

5.14E-24

7.10E-23

1.19E-23

5686/5688/5682/5710/5707/5718/5695/5689/5691/5702


Table 6 is not available with this version