Transcriptomic Changes Highly Similar to Alzheimer’s Disease Are Observed in a Subpopulation of Individuals During Normal Brain Aging

Shouneng Peng1,2,3, Lu Zeng1,2,3, Jean‐Vianney Haure‐Mirande4, Minghui Wang1,2,3, Derek M. Huffman5,6,7, Vahram Haroutunian8,9,10, Michelle E. Ehrlich1,4,11, Bin Zhang1,2,3, Zhidong Tu1,2,3
1Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
2Icahn Institute for Data Science and Genomic Technology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
3Institute of Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
4Department of Neurology, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
5Department of Medicine, Albert Einstein College of Medicine, New York City, NY, United States
6Department of Molecular Pharmacology, Albert Einstein College of Medicine, New York City, NY, United States
7Institute for Aging Research, Albert Einstein College of Medicine, New York City, NY, United States
8Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
9Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
10Mental Illness Research, Education and Clinical Center (MIRECC), James J. Peters Department of Veterans Affairs Medical Center, Bronx, NY, United States
11Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York City, NY, United States

Tóm tắt

Aging is a major risk factor for late-onset Alzheimer’s disease (LOAD). How aging contributes to the development of LOAD remains elusive. In this study, we examined multiple large-scale transcriptomic datasets from both normal aging and LOAD brains to understand the molecular interconnection between aging and LOAD. We found that shared gene expression changes between aging and LOAD are mostly seen in the hippocampal and several cortical regions. In the hippocampus, the expression of phosphoprotein, alternative splicing and cytoskeleton genes are commonly changed in both aging and AD, while synapse, ion transport, and synaptic vesicle genes are commonly down-regulated. Aging-specific changes are associated with acetylation and methylation, while LOAD-specific changes are more related to glycoprotein (both up- and down-regulations), inflammatory response (up-regulation), myelin sheath and lipoprotein (down-regulation). We also found that normal aging brain transcriptomes from relatively young donors (45–70 years old) clustered into several subgroups and some subgroups showed gene expression changes highly similar to those seen in LOAD brains. Using brain transcriptomic datasets from another cohort of older individuals (>70 years), we found that samples from cognitively normal older individuals clustered with the “healthy aging” subgroup while AD samples mainly clustered with the “AD similar” subgroups. This may imply that individuals in the healthy aging subgroup will likely remain cognitively normal when they become older and vice versa. In summary, our results suggest that on the transcriptome level, aging and LOAD have strong interconnections in some brain regions in a subpopulation of cognitively normal aging individuals. This supports the theory that the initiation of LOAD occurs decades earlier than the manifestation of clinical phenotype and it may be essential to closely study the “normal brain aging” to identify the very early molecular events that may lead to LOAD development.

Từ khóa


Tài liệu tham khảo

2021, 2021 Alzheimer’s disease facts and figures., Alzheimers Dement., 17, 327, 10.1002/alz.12328

Annese, 2018, Whole transcriptome profiling of Late-Onset Alzheimer’s disease patients provides insights into the molecular changes involved in the disease., Sci. Rep., 8, 4282, 10.1038/s41598-018-22701-2

Berchtold, 2013, Synaptic genes are extensively downregulated across multiple brain regions in normal human aging and Alzheimer’s disease., Neurobiol. Aging, 34, 1653, 10.1016/j.neurobiolaging.2012.11.024

Bordner, 2011, Parallel declines in cognition, motivation, and locomotion in aging mice: association with immune gene upregulation in the medial prefrontal cortex., Exp. Gerontol., 46, 643, 10.1016/j.exger.2011.03.003

Canty, 1994, Lecithin and choline in human health and disease., Nutr. Rev., 52, 327, 10.1111/j.1753-4887.1994.tb01357.x

Caselli, 2009, Longitudinal modeling of age-related memory decline and the APOE epsilon4 effect., N. Engl. J. Med., 361, 255, 10.1056/NEJMoa0809437

Consortium, 2015, Human genomics. The genotype-tissue expression (GTEx) pilot analysis: multitissue gene regulation in humans., Science, 348, 648, 10.1126/science.1262110

Cribbs, 2012, Extensive innate immune gene activation accompanies brain aging, increasing vulnerability to cognitive decline and neurodegeneration: a microarray study., J. Neuroinflammation, 9, 179, 10.1186/1742-2094-9-179

Ferreira, 2020, Biological subtypes of Alzheimer disease: a systematic review and meta-analysis., Neurology, 94, 436, 10.1212/WNL.0000000000009058

Herrup, 2010, Reimagining Alzheimer’s disease–an age-based hypothesis., J. Neurosci., 30, 16755, 10.1523/JNEUROSCI.4521-10.2010

Hodes, 2016, Accelerating medicines partnership: Alzheimer’s disease (AMP-AD) knowledge portal aids Alzheimer’s drug discovery through open data sharing., Expert Opin. Ther. Targets, 20, 389, 10.1517/14728222.2016.1135132

Hopperton, 2018, Markers of microglia in post-mortem brain samples from patients with Alzheimer’s disease: a systematic review., Mol. Psychiatry, 23, 177, 10.1038/mp.2017.246

Huang, 2009, Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists., Nucleic Acids Res., 37, 1, 10.1093/nar/gkn923

Jack, 2015, Age, sex, and APOE epsilon4 effects on memory, brain structure, and beta-amyloid across the adult life span., JAMA Neurol., 72, 511, 10.1001/jamaneurol.2014.4821

Jacquet, 2009, FoxJ1-dependent gene expression is required for differentiation of radial glia into ependymal cells and a subset of astrocytes in the postnatal brain., Development, 136, 4021, 10.1242/dev.041129

Janz, 1998, SVOP, an evolutionarily conserved synaptic vesicle protein, suggests novel transport functions of synaptic vesicles., J. Neurosci., 18, 9269, 10.1523/JNEUROSCI.18-22-09269.1998

Kiltschewskij, 2017, Post-transcriptional mechanisms of neuronal translational control in synaptic plasticity, Synaptic Plasticity, 15

Koivisto, 1995, Prevalence of age-associated memory impairment in a randomly selected population from eastern Finland., Neurology, 45, 741, 10.1212/wnl.45.4.741

Lanke, 2018, Integrative analysis of hippocampus gene expression profiles identifies network alterations in aging and Alzheimer’s disease., Front. Aging Neurosci., 10, 153, 10.3389/fnagi.2018.00153

Lau, 2020, Single-nucleus transcriptome analysis reveals dysregulation of angiogenic endothelial cells and neuroprotective glia in Alzheimer’s disease., Proc. Natl. Acad. Sci. U.S.A., 117, 25800, 10.1073/pnas.2008762117

Law, 2014, voom: precision weights unlock linear model analysis tools for RNA-seq read counts., Genome Biol., 15, R29, 10.1186/gb-2014-15-2-r29

Licata, 2020, SIGNOR 2.0, the SIGnaling network open resource 2.0: 2019 update., Nucleic Acids Res., 48, D504, 10.1093/nar/gkz949

Lin, 2004, Modulation of Th1 activation and inflammation by the NF-kappaB repressor Foxj1., Science, 303, 1017, 10.1126/science.1093889

Mahfouz, 2015, Visualizing the spatial gene expression organization in the brain through non-linear similarity embeddings., Methods, 73, 79, 10.1016/j.ymeth.2014.10.004

Mastroeni, 2017, Nuclear but not mitochondrial-encoded oxidative phosphorylation genes are altered in aging, mild cognitive impairment, and Alzheimer’s disease., Alzheimers Dement., 13, 510, 10.1016/j.jalz.2016.09.003

Mathys, 2019, Single-cell transcriptomic analysis of Alzheimer’s disease., Nature, 570, 332, 10.1038/s41586-019-1195-2

Millar, 2007, Tissue and organ donation for research in forensic pathology: the MRC sudden death brain and tissue bank., J. Pathol., 213, 369, 10.1002/path.2247

Mostafavi, 2018, A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer’s disease., Nat. Neurosci., 21, 811, 10.1038/s41593-018-0154-9

Murtagh, 2014, Ward’s hierarchical agglomerative clustering method: which algorithms implement Ward’s criterion?, J. Classif., 31, 274, 10.1007/s00357-014-9161-z

Nakamura, 2001, Progressive brain dysfunction following intracerebroventricular infusion of beta(1-42)-amyloid peptide., Brain Res., 912, 128, 10.1016/s0006-8993(01)02704-4

Neff, 2021, Molecular subtyping of Alzheimer’s disease using RNA sequencing data reveals novel mechanisms and targets., Sci. Adv., 7, eabb5398, 10.1126/sciadv.abb5398

Paik, 2019, Somatostatin maintains permeability and integrity of blood-brain barrier in beta-amyloid induced toxicity., Mol. Neurobiol., 56, 292, 10.1007/s12035-018-1045-5

Patrick, 2020, Deconvolving the contributions of cell-type heterogeneity on cortical gene expression., PLoS Comput. Biol., 16, e1008120, 10.1371/journal.pcbi.1008120

Reichwald, 2009, Expression of complement system components during aging and amyloid deposition in APP transgenic mice., J. Neuroinflammation, 6, 35, 10.1186/1742-2094-6-35

Robinson, 2010, edgeR: a Bioconductor package for differential expression analysis of digital gene expression data., Bioinformatics, 26, 139, 10.1093/bioinformatics/btp616

Sherman, 2009, Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources., Nat. Protoc., 4, 44, 10.1038/nprot.2008.211

Solarski, 2018, Somatostatin in Alzheimer’s disease: a new role for an old player., Prion, 12, 1, 10.1080/19336896.2017.1405207

Trabzuni, 2011, Quality control parameters on a large dataset of regionally dissected human control brains for whole genome expression studies., J. Neurochem., 119, 275, 10.1111/j.1471-4159.2011.07432.x

van Rooij, 2019, Hippocampal transcriptome profiling combined with protein-protein interaction analysis elucidates Alzheimer’s disease pathways and genes., Neurobiol. Aging, 74, 225, 10.1016/j.neurobiolaging.2018.10.023

Verbitsky, 2004, Altered hippocampal transcript profile accompanies an age-related spatial memory deficit in mice., Learn. Mem., 11, 253, 10.1101/lm.68204

Wang, 2018, The Mount Sinai cohort of large-scale genomic, transcriptomic and proteomic data in Alzheimer’s disease., Sci. Data, 5, 180185, 10.1038/sdata.2018.185

Wang, 2016, Integrative network analysis of nineteen brain regions identifies molecular signatures and networks underlying selective regional vulnerability to Alzheimer’s disease., Genome Med., 8, 104, 10.1186/s13073-016-0355-3

Yang, 2015, Synchronized age-related gene expression changes across multiple tissues in human and the link to complex diseases., Sci. Rep., 5, 15145, 10.1038/srep15145

Yao, 2009, SVOP is a nucleotide binding protein., PLoS One, 4, e5315, 10.1371/journal.pone.0005315

Yu, 2008, Foxj1 transcription factors are master regulators of the motile ciliogenic program., Nat. Genet., 40, 1445, 10.1038/ng.263

Zarow, 2005, Correlates of hippocampal neuron number in Alzheimer’s disease and ischemic vascular dementia., Ann. Neurol., 57, 896, 10.1002/ana.20503

Zeisel, 2007, Gene response elements, genetic polymorphisms and epigenetics influence the human dietary requirement for choline., IUBMB Life, 59, 380, 10.1080/15216540701468954

Zeng, 2020, Transcriptome analysis reveals the difference between “healthy” and “common” aging and their connection with age-related diseases., Aging Cell, 19, e13121, 10.1111/acel.13121

Zhang, 2016, Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse., Neuron, 89, 37, 10.1016/j.neuron.2015.11.013

Zhong, 2013, Digital sorting of complex tissues for cell type-specific gene expression profiles., BMC Bioinformatics, 14, 89, 10.1186/1471-2105-14-89

Zhou, 2019, NetworkAnalyst 3.0: a visual analytics platform for comprehensive gene expression profiling and meta-analysis., Nucleic Acids Res., 47, W234, 10.1093/nar/gkz240