Skip to main content
Top
Published in: BMC Neurology 1/2021

01-12-2021 | Alzheimer's Disease | Research

Cell type-specific potential pathogenic genes and functional pathways in Alzheimer’s Disease

Authors: Xiao-Lan Wang, Lianjian Li

Published in: BMC Neurology | Issue 1/2021

Login to get access

Abstract

Background

Alzheimer's disease (AD) is a pervasive age-related and highly heritable neurodegenerative disorder but has no effective therapy. The complex cellular microenvironment in the AD brain impedes our understanding of pathogenesis. Thus, a comprehensive investigation of cell type-specific responses in AD is crucial to provide precise molecular and cellular targets for therapeutic development.

Methods

Here, we integrated analyzed 4,441 differentially expressed genes (DEGs) that were identified from 263,370 single-cells in cortex samples by single-nucleus RNA sequencing (snRNA-seq) between 42 AD-pathology subjects and 39 normal controls within 3 studies. DEGs were analyzed in microglia, astrocytes, oligodendrocytes, excitatory neurons, inhibitory neurons, and endothelial cells, respectively. In each cell type, we identified both common DEGs which were observed in all 3 studies, and overlapping DEGs which have been seen in at least 2 studies. Firstly, we showed the common DEGs expression and explained the biological functions by comparing with existing literature or multil-omics signaling pathways knowledgebase. We then determined the significant modules and hub genes, and explored the biological processes using the overlapping DEGs. Finally, we identified the common and distinct dysregulated pathways using overall DEGs and overlapping DEGs in a cell type-specific manner.

Results

Up-regulated LINGO1 has been seen in both oligodendrocytes and excitatory neurons across 3 studies. Interestingly, genes enriched in the mitochondrial module were up-regulated across all cell types, which indicates mitochondrial dysfunction in the AD brain. The estrogen signaling pathway seems to be the most common pathway that is disrupted in AD.

Conclusion

Together, these analyses provide detailed information of cell type-specific and overall transcriptional changes and pathways underlying the human AD-pathology. These findings may provide important insights for drug development to tackle this disease.
Appendix
Available only for authorised users
Literature
21.
37.
45.
go back to reference Rangaraju S, Dammer EB, Raza SA, Rathakrishnan P, Xiao HL, Gao TW, et al. Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer's disease. Mol Neurodegener. 2018;13:ARTN 24. https://doi.org/10.1186/s13024-018-0254-8 Rangaraju S, Dammer EB, Raza SA, Rathakrishnan P, Xiao HL, Gao TW, et al. Identification and therapeutic modulation of a pro-inflammatory subset of disease-associated-microglia in Alzheimer's disease. Mol Neurodegener. 2018;13:ARTN 24. https://​doi.​org/​10.​1186/​s13024-018-0254-8
49.
go back to reference Zhou XP, Chen Y, Mok KY, Kwok TCY, Mok VCT, Guo QH, et al. Non-coding variability at the APOE locus Communications. 2019; 10:Artn 3310. doi: 10.38/S41467-019-10945-Z Zhou XP, Chen Y, Mok KY, Kwok TCY, Mok VCT, Guo QH, et al. Non-coding variability at the APOE locus Communications. 2019; 10:Artn 3310. doi: 10.38/S41467-019-10945-Z
51.
54.
64.
go back to reference Kim JH, Franck J, Kang T, Heinsen H, Ravid R, Ferrer I, et al. Proteome-wide characterization of signalling interactions in the hippocampal CA4/DG subfield of patients with Alzheimer's disease. Scientific Reports. 2015;5:Artn 11138. https://doi.org/10.1038/Srep11138 Kim JH, Franck J, Kang T, Heinsen H, Ravid R, Ferrer I, et al. Proteome-wide characterization of signalling interactions in the hippocampal CA4/DG subfield of patients with Alzheimer's disease. Scientific Reports. 2015;5:Artn 11138. https://​doi.​org/​10.​1038/​Srep11138
66.
go back to reference Shigemizu D, Akiyama S, Asanomi Y, Boroevich KA, Sharma A, Tsunoda T, et al. Risk prediction models for dementia constructed by supervised principal component analysis using miRNA expression data. Communications Biology. 2019;2:ARTN 77. https://doi.org/10.1038/s42003-019-0324-7 Shigemizu D, Akiyama S, Asanomi Y, Boroevich KA, Sharma A, Tsunoda T, et al. Risk prediction models for dementia constructed by supervised principal component analysis using miRNA expression data. Communications Biology. 2019;2:ARTN 77. https://​doi.​org/​10.​1038/​s42003-019-0324-7
72.
78.
go back to reference Blennow K, Bogdanovic N, Alafuzoff I, Ekman R, Davidsson P. Synaptic pathology in Alzheimer's disease: Relation to severity of dementia, but not to senile plaques, neurofibrillary tangles, or the ApoE4 allele. J Neural Transmission. 1996;103:603. https://doi.org/10.1007/Bf01273157 Blennow K, Bogdanovic N, Alafuzoff I, Ekman R, Davidsson P. Synaptic pathology in Alzheimer's disease: Relation to severity of dementia, but not to senile plaques, neurofibrillary tangles, or the ApoE4 allele. J Neural Transmission. 1996;103:603. https://​doi.​org/​10.​1007/​Bf01273157
90.
95.
go back to reference Frisard M, Ravussin E. Energy metabolism and oxidative stress - Impact on the metabolic syndrome and the aging process. Endocrine. 2006; 29:27. doi: Doi 10.1385/Endo:29:1:27 Frisard M, Ravussin E. Energy metabolism and oxidative stress - Impact on the metabolic syndrome and the aging process. Endocrine. 2006; 29:27. doi: Doi 10.1385/Endo:29:1:27
100.
go back to reference Konishi H, Okamoto T, Hara Y, Komine O, Tamada H, Maeda M, et al. Astrocytic phagocytosis is a compensatory mechanism for microglial dysfunction. Embo J. (2020) 39. doi: Konishi H, Okamoto T, Hara Y, Komine O, Tamada H, Maeda M, et al. Astrocytic phagocytosis is a compensatory mechanism for microglial dysfunction. Embo J. (2020) 39. doi:
102.
go back to reference Gabbouj S, Ryhanen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al. Altered Insulin Signaling in Alzheimer's Disease Brain - Special Emphasis on PI3K-Akt Pathway. Front Neurosci-Switz. (2019) 13. doi: Artn 62910.3389/Fnins.2019.00629 Gabbouj S, Ryhanen S, Marttinen M, Wittrahm R, Takalo M, Kemppainen S, et al. Altered Insulin Signaling in Alzheimer's Disease Brain - Special Emphasis on PI3K-Akt Pathway. Front Neurosci-Switz. (2019) 13. doi: Artn 62910.3389/Fnins.2019.00629
103.
go back to reference Cianciulli A, Porro C, Calvello R, Trotta T, Lofrumento DD, Panaro MA. Microglia Mediated Neuroinflammation: Focus on PI3K Modulation. Biomolecules. (2020) 10. doi: Artn 13710.3390/Biom10010137 Cianciulli A, Porro C, Calvello R, Trotta T, Lofrumento DD, Panaro MA. Microglia Mediated Neuroinflammation: Focus on PI3K Modulation. Biomolecules. (2020) 10. doi: Artn 13710.3390/Biom10010137
106.
go back to reference Lam D, Enright HA, Cadena J, Peters SKG, Sales AP, Osburn JJ, et al. Tissue-specific extracellular matrix accelerates the formation of neural networks and communities in a neuron-glia co-culture on a multi-electrode array. Scientific Reports. 2019;9:Artn 4159. https://doi.org/10.1038/S41598-019-40128-1 Lam D, Enright HA, Cadena J, Peters SKG, Sales AP, Osburn JJ, et al. Tissue-specific extracellular matrix accelerates the formation of neural networks and communities in a neuron-glia co-culture on a multi-electrode array. Scientific Reports. 2019;9:Artn 4159. https://​doi.​org/​10.​1038/​S41598-019-40128-1
Metadata
Title
Cell type-specific potential pathogenic genes and functional pathways in Alzheimer’s Disease
Authors
Xiao-Lan Wang
Lianjian Li
Publication date
01-12-2021
Publisher
BioMed Central
Published in
BMC Neurology / Issue 1/2021
Electronic ISSN: 1471-2377
DOI
https://doi.org/10.1186/s12883-021-02407-1

Other articles of this Issue 1/2021

BMC Neurology 1/2021 Go to the issue