Abstract
Advances and reduction of costs in various sequencing technologies allow for a closer look at variations present in the non-coding regions of the human genome. Correlating non-coding variants with large-scale multi-omic data holds the promise not only of a better understanding of likely causal connections between non-coding DNA and expression of traits but also identifying potential disease-modifying medicines. Genome–phenome association studies have created large datasets of DNA variants that are associated with multiple traits or diseases, such as Alzheimer’s disease; yet, the functional consequences of variants, in particular of non-coding variants, remain largely unknown. Recent advances in functional genomics and computational approaches have led to the identification of potential roles of DNA variants, such as various quantitative trait locus (xQTL) techniques. Multi-omic assays and analytic approaches toward xQTL have identified links between genetic loci and human transcriptomic, epigenomic, proteomic and metabolomic data. In this review, we first discuss the recent development of xQTL from multi-omic findings. We then highlight multimodal analysis of xQTL and genetic data for identification of risk genes and drug targets using Alzheimer’s disease as an example. We finally discuss challenges and future research directions (e.g. artificial intelligence) for annotation of non-coding variants in complex diseases.
Introduction
Estimates of the percentage of the human genome that is ultimately translated into protein vary but are generally believed to be within the range of 1–2% of the entire human genome (1). The rest of the human genome is classified into a wide variety of categories, such as regulatory elements, promoters (2), non-coding genes (3), introns (4) and sections relating to chromosomal structure, activity or segregation (5). Most previous research on DNA variants associated with diseases and traits have focused on the exome part of the genome due to cost, speed and the belief that the coding regions hold the majority of functional variation. However, falling costs of next-generation sequencing technologies allows for broader-scope studies of the whole genome and with more participants than was previously feasible. Apart from teasing out disease associations with genes that do have known functions, a non-insignificant portion of disease-associated genetic risk loci that have been identified are located in non-coding sections of the genome, whose functions are poorly understood.
The influence that non-coding DNA variants identified in whole-genome sequencing (WGS) and genome-wide association studies (GWAS) have on gene expression, post-transcriptional modification and regulation of protein activity suggests that study of those regions can be valuable in finding new associations between DNA variants and disease phenotypes (6–8). Recent advances in functional genomics and computational approaches have led to the identification of potential roles of DNA variants, such as various quantitative trait loci (xQTL), for disease risk gene and drug target identification. However, the predisposition to complex diseases [including Alzheimer’s disease (AD)] involves a complex, polygenic and pleiotropic genetic architecture. Systematic investigation of DNA variant effects on xQTL by unique integration of human genome, transcriptome, epigenome, proteome and metabolome are highly valuable for research and healthcare applications.
Characteristics of xQTL
A QTL is a relationship between a genomic locus whose variation is associated with a quantitative phenotype. xQTL refers to a range of techniques that correlate genetic variants with multi-omic data, for the purposes of connecting genomic variants with phenotypic traits. While expression quantitative trait loci (eQTL) is one of the more common xQTL analyses performed, epigenomic, transcriptomic, proteomic, metabolomics and other QTL analyses present the need for analyzing multi-omic data types and drawing correlations to phenotypes. We briefly highlight several xQTLs below, including eQTL, protein expression QTL (pQTL), histone-QTL (hQTL), splicing QTL (sQTL), methylation QTL (meQTL) and metabolite QTL (mQTL). Figure 1 illustrates a diagram of several types of xQTL.
Figure 1.
Diagrammatic illustration of various quantitative trait loci (xQTL). The discussed xQTL include gene expression quantitative trait loci (eQTL), protein expression QTL (pQTL), splicing QTL (sQTL), histone QTL (hQTL), methylation QTL (meQTL) and other types of QTL [such as metabolite QTL (mQTL)]. These xQTL can be identified from various transcriptomic (RNA-seq), epigenomic (DNA methylation-seq and ChiP-seq, ATAC-seq) and other types of multi-omic assays.
Expression quantitative trait loci
eQTL refers to identifying genetic loci that are connected to levels of expression of DNA products (9). eQTL relies on the quality of multiple techniques: DNA sequencing, quantification of mRNA expression (10,11), and the accuracy of data analysis methods (12,13). The cheapening of DNA sequencing technology over time, as well as advances of mRNA-sequencing technologies, have contributed to the feasibility of conducting large-scale eQTL studies with input from thousands of people. eQTL’s potential is not restricted to comparison of traits in populations, but is also highly useful in analyzing transcriptome data from various tissues and cell types (14,15). An example is the Genotype-Tissue Expression (GTEx) project (16,17), which aims to catalog the tissue transcriptome in connection with genomic variants throughout the body. eQTL also has potential in researching transcriptional changes in response to drug treatment (18,19).
Protein expression quantitative trait loci
While quantitation of mRNA expression is very often used to indirectly measure the expression of proteins overall within cells or tissues, eQTL data may not accurately reflect protein levels due to post-transcriptional regulation and degradation (11,20). Unlike pQTL, eQTL may not capture tissue-specific differences, complex pathways or protein interactions (21–23). pQTL has the potential advantage of being better able to link specific proteins to expression of a disease or trait than eQTL alone (24), and thus, is highly advantageous in the process of functional annotation and target identification during drug discovery and development (25). pQTL also is not restricted to cellular analysis but can test a variety of biological samples and tissues, such as blood plasma and cerebrospinal fluid (CSF) (24,26).
Histone-QTL
Transcription of a DNA segment is generally negatively correlated to the amount of histones bound to that segment (27). Histone–DNA affinity is mainly mediated by electrostatic attraction between lysine and arginine on histones and the DNA (28). The activity and binding of histones can be modified through several mechanisms that can add/remove charges on histone proteins or alter their conformation (29). Besides altering histone binding itself, modification of histone proteins can also affect transcription by presenting/hiding epitopes to transcription factors (30,31). Histone modifications are heavily involved in regulation of aging, responses to environmental stressors and diseases such as cancer (32–34). Polymorphisms in histone acetylation and methylation regulating enzymes have been implicated in a broad range of neurodevelopmental disorders (37). Histone binding/modifications are one of the two major branches of epigenetics, the other being DNA methylation (35,36). Because of histone’s influence on DNA transcription, hQTL studies can research interactions between histones and DNA variants in two ways: (i) researching genomic regions and variants associated with variations in histone modifications/binding themselves (37) and (ii) analyzing locations of heavy histone binding/modification as loci themselves, and correlating the impact of histones with eQTL and phenotypic data (38,39).
Splicing QTL
sQTL refers to techniques that correlate genomic variants associated with alternative splicing and that look for causal connections between alternative splicing and trait expression. Alternative splicing of pre-mRNAs to produce different variants of the same protein is extremely common, with 92–94% of all genes in humans being subject to alternative splicing, at least occasionally (40). Patterns of alternative splicing is highly tissue-specific (41) and are important for regulation of cell differentiation and embryonic development (42). Within the brain, splicing is highly regulated, especially during embryonic development (43). Alternative splicing can directly contribute towards disease, such as alternative splicing of tau protein isoforms in AD and other tauopathies (44,45), or by impairment of the spliceosomal complex itself (46,47). In Takata et al. (46), a high percentage of splicing loci was shown to be associated with previously identified genetic loci associated with neurological disorders, and dysregulation of splicing is heavily associated with AD, schizophrenia and autism spectrum disorder in multiple studies (47–50).
DNA methylation QTL
An additional dimension of DNA sequencing is methyl sequencing, or methyl-seq, which identifies the presence of methyl groups on adenine and cytosine bases on DNA. DNA methylation is an important pathway for modifying gene expression, displaying a variety of effects on up/downregulating transcriptional activity and alternative splicing dependent on the region(s) of the genome that are methylated. DNA methylation’s role in human disease has mainly been investigated in relation to cancer (51,52), but meQTL studies have identified methylation loci that are involved in immune system functioning (53) and AD (54). As with hQTL, researchers can take the same two approaches discussed previously and apply them to studies on meQTL (55,56).
Metabolite QTL
Metabolites are chemical compounds that are produced by a cell or organism as it goes through metabolic processes. The metabolome, like other qualities of an organism, can be profiled with a variety of technologies (57). Metabolites, unlike RNAs or proteins, do not have a direct relationship with DNA variants, being the result of the interactions of many cellular processes. However, metabolites can and have been correlated with genomic variants in mQTL studies (58,59). mQTL can also be coupled with investigations into the microbiome (microbiome QTL), in order to analyze relationships between the microbial composition of a tissue, metabolites produced by resident microbes, genomic variations within the host and resulting effects on the phenotype of the host (60–62). Thus, mQTL offers a powerful tool to investigate gene–environment interactions for non-coding variants.
Resources of human genome-sequencing and multi-omic data
High-throughput DNA/RNA sequencing technologies have generated a large amount of genomic data in multiple national/international genome sequencing projects, such as UK Biobank (63,64), TOPMed (65), All of US (66), Biobank Japan (67,68) and the GWAS Catalog (69). All of these databases tend to focus on diverse populations or different diseases, but share the similarities of being data repositories for genomic and health information for large number of participants. A list of these databases can be located in Table 1 of this article.
Table 1.
List of data resources for human genome sequencing and functional genomic studies (including xQTL). Resources of human genome-sequencing and multi-omic data
Name | Description | Website | Ref. |
---|---|---|---|
Human genome-sequencing | |||
Alzheimer’s Disease Sequencing Project (ADSP) | Identifying genomic variants that affect the risk of developing AD | https://www.niagads.org/adsp/content/home | (73) |
Alzheimer’s Disease Neuroimaging Initiative (ADNI) | Use of brain MRI and PET scans, SNP sequencing, and biospecimens to diagnose and intervene in AD development | https://adni.loni.usc.edu/ | (74) |
The Encyclopedia of DNA Elements (ENCODE) | Functional annotation of elements in the human genome | https://www.encodeproject.org/ | (79) |
NIAGADS | Repository of genomic sequencing data to study late-onset AD | https://www.niagads.org/adsp/ | (75) |
Functional Annotation Of The Mammalian Genome (FANTOM5) | Transcriptomic analysis of various cell types to understand how transcriptional control and gene expression occur | https://fantom.gsc.riken.jp/5/ | (83) |
UK BioBank | Collecting DNA variants and health information from participants in the UK | https://www.ukbiobank.ac.uk/ | (64) |
Trans-Omics for Precision Medicine (TOPMed) Program | Collecting genomics heart, lung, blood and sleep disorders | https://topmed.nhlbi.nih.gov/ | (65) |
GWAS Catalog | To serve as a database for GWAS data in general | https://www.ebi.ac.uk/gwas/ | (69) |
All of Us | Collecting diverse health data from more than 1 million participants within the US | https://allofus.nih.gov/ | (66) |
BioBank Japan | Collecting genomic, biospecimen and health data from Japanese participants | https://biobankjp.org/en/ | (68) |
Multi-omic resources | |||
The Genotype-Tissue Expression (GTEx) project | To collate together gene expression data across the various tissue types and organs present in the human body | https://gtexportal.org/home/ | (16) |
AD knowledge portal | Mapping of human proteins within the body | https://adknowledgeportal.synapse.org/ | (132) |
alzGPS | Integrating multi-omics, clinical trials and drug target data for AD drug discovery/repurposing | https://alzgps.lerner.ccf.org/ | (77) |
The Alzheimer’s Cell Atlas (TACA) | Correlation of AD pathobiology with single-cell RNA-Seq from AD patients and model mice | https://taca.lerner.ccf.org/ | (78) |
For example, UK Biobank, which follows approximately 500 000 residents of the UK, has generated the whole-genome sequences of 150 119 individuals and identified more than 600 million sequence variants (70). TOPMed, which is sponsored by NIH, focuses on multi-omic studies on heart, lung, blood and sleep disorders, and has sequenced more than 53 000 whole genomes with 410 million sequence variants identified (65). All of US, also operated by NIH, focuses on collecting electronic health records, surveys and biospecimens to reflect the diverse population of the US, and has gathered data from 370 000 participants (accessed July 24, 2022) (71, 72). BioBank Japan collected medical records and tissue and DNA samples from approximately 200 000 participants in Japan enrolled from 2003 to 2008 (68). The GWAS Catalog, run cooperatively by the National Human Genome Research Institute (NHGRI) and the European Bioinformatics Institute (EBI), does not run data collection by themselves, but does collect and organize GWAS data from a variety of other sources (69).
For AD, there are several specific genome-sequencing and multi-omic resources, including the Alzheimer’s Disease Sequencing Project (ADSP) (73), the Alzheimer’s Disease Neuroimaging Initiative (ADNI) (74) and the Genetics of Alzheimer’s Disease Data Storage Site (NIAGADS) (75). NIAGADS is a national genetic and genomic data repository for AD research that to date has collected 90 datasets and 144 242 samples (accessed July 19, 2022). The ADSP, launched in 2012, has sequenced and analyzed genomes to identify a wide range of both risk and protective genetic variants in AD, including whole-exome sequencing (WES) data from nearly 20 000 samples, as well as WGS data from nearly 17 000 samples (73). The AD Knowledge Portal (76) contains upwards of 100 000 data files from over 80 studies of people with AD and related model organisms. This resource provides rich xQTL, genomic, epigenomic, transcriptomic, proteomic and metabolomic data from thousands of human brains. A few projects play an outsized role in being sources for xQTL data involving the brain or cognitive functions: the ROSMAP project, the Mount Sinai Brain Bank (MSBB) and the Mayo RNA-Seq project (77–79). The ROSMAP study, which is the combined data from the Religious Orders Study and the Memory and Aging Project, recruits religious members (ROS) and people from the Greater Chicago Area (MAP), collect data from cognitive tests and correlate them to genomic variants and other –omics data. MSBB combines results from cognitive tests and correlates them to Braak-Staged brains and –omics data from participants. Mayo RNA-Seq focuses mainly on transcriptomic data from banked brains with AD and other neurodegenerative disorders. Several recent multi-omic databases, such as alzGPS (77) and The Alzheimer’s Cell Atlas (78) have leveraged large-scale multi-omic data available for target identification.
Several commonly used functional genomic data resources include ENCODE (79,80), GTEx (16,81) and FANTOM5 (82,83). ENCODE aims to functionally annotate the entire human and mouse genomes, and thus it is focused on data analysis of other studies, with over 5000 datasets (79). GTEx specifically aims to create an atlas of expression data based on tissue. In its most recent official data release (GTEx Analysis V8), the GTEx project has obtained 17 382 total samples and 15 201 eQTL analyzed samples, from 948 and 838 donors, respectively. FANTOM5, which is a project of the FANTOM consortium, collects transcriptome, lncRNA and miRNA data from a wide variety of human and mouse tissues and cell lines to study transcriptional regulation (84), with over 1000 human and mouse samples.
xQTL-based annotation of non-coding variants in AD
While non-coding DNA, by its very definition, is not transcribed into proteins, non-coding DNA serves a variety of other functions essential to functioning and regulation of gene expression (85,86). The functions of these non-coding genes include: the transcription of regulatory RNAs from non-coding genes (87–89), promoters and regulatory elements that control gene transcription (90,91), introns and structural functions, such as centromere activity (5,92). With this in mind, the effects of non-coding DNA variants fall generally under either affecting levels of gene expression, affecting post-transcriptional modification of mRNA transcripts or changing the sequence of non-coding RNAs (6,7,93). For analyzing these effects, eQTL, pQTL and splice-QTL are of great value.
AD is one of the most common forms of dementia diagnosed in the elderly (94), characterized by amyloid-beta plaques and phosphorylated tau tangles. AD can generally be classified into two types, based on disease etiology: Familial Alzheimer’s Disease (FAD), which is characterized by mutations in key AD-related genes (PSEN1, PSEN2 and APP) and an early age of onset (95) and Late-Onset Alzheimer’s Disease (LOAD), which is dependent on complex interactions between genetic factors, health conditions and lifestyle factors. While many WGS/WES association studies have been conducted on patients with AD and other cognitive impairments, fewer genes have been conclusively linked to an increased risk of developing LOAD, with the notable exception of the APOE4 variant of APOE (96). Variants in proximity to APOE, which is itself a well-known AD risk gene, have been found in Blue et al. (97) and Zhou et al. (98), from data obtained from two different groups of participants. Other non-coding variants have found variants in proximity to AD risk genes observed in other studies (99), such as BIN1 (100) and CD33 (101).
In Raj et al. (48), which studied alterations in splicing in the prefrontal cortex of 450 subjects, used LeafCutter software to estimate excised introns in mRNAs (102). In Raj et al., the RNA-Seq results were also compared against data from MSBB and iPSC lines in a replication analysis. The data were compared with genomic data from the participants to analyze associations between sQTLs and AD-related genes. Yang et al. (24) performed pQTL studies on CSF, brain tissue and blood from participants with or without AD. The researchers used an aptamer platform, and discovered that cis-QTLs (within close proximity of a protein-coding gene), tend to affect all sample tissues from a subject, while trans-QTLs tend to be more tissue-specific. The researchers also replicated their findings against multiple pre-existing datasets. Zhao et al. (103) conducted a meta-analysis on five GWAS datasets, three eQTL datasets and three mQTL datasets, obtained mostly from brain tissue, to analyze AD-related genes. The researchers used Summary data-based Mendelian randomization to compare the different datasets, and found significant concordance between all three types of datasets in terms of AD-related genes identified. In Wang et al. (104), the researchers focused on DNA methylation in peripheral blood in the context of MCI and AD, and found a reduction in promoter methylation in the PM20D1 locus in MCI and AD patients. Similarly, Hillary et al. (105) also focused on DNA methylation, as well as pQTL, correlating the blood levels of 18 AD-associated proteins to variations in 19 CpG sites, and pQTL associated with increased expression of TREM2. Ng et al. (106) take a broader approach, eQTL, hQTL and meQTL techniques to analyze the dorsolateral prefrontal cortex of 411 participants to map the epigenome and transcriptome, and identify new susceptibility genes for schizophrenia and bipolar disorder. Finally, Novikova et al. (107) studied myeloid cell and microglial enhancers with hQTL. In all of these studies, certain commonalities regarding the cell types or genes affected in AD or MCI can be noted, regardless of the tissue type or xQTL studied in the particular article.
There are several potential limitations that can occur with xQTL and GWAS studies. Variants may be present at very low allele frequencies within the population or may be de novo mutations present only in the patient or their family, which might mean that statistical analyses might not have enough power to make a meaningful correlation (108,109). Synergistic effects among different variants is also a factor that needs to be considered during analysis (110,111). SNP/variant frequencies can differ markedly between demographic groups being analyzed (112,113), which can create inaccuracies when applying GWAS results to populations from ancestries other than the population sampled in the study (113–115). An additional challenge when analyzing and annotating variants is linkage disequilibrium due to co-inheritance of sections of the genome during homologous recombination. As a result, techniques such as massively parallel reporter assays are often need to be used to discern causal variants (116).
Discussion and Future Directions
Investigating the polygenic influences on the majority of human illnesses and traits requires the accurate collection and analysis of increasingly larger sets of data generated from patients. While the advances in xQTL technologies have been beneficial in shedding light on biological pathways, sifting through the massive amounts of data to identify meaningful results requires taking care to avoid bias in participant selection, as well as the aid of a variety of computational algorithms. xQTL analysis is a powerful tool for discovering biological pathways, annotation of non-coding variants and potential therapeutic treatments. The main advantage of xQTL studies is the ability to collect and correlate multi-omics together in order to identify likely causal risk genes and drug targets. While examining genomic, transcriptomic or other data alone in comparison with patient data is valuable, integrative analysis allows for information to be collected about the effects of genomic and other variation not just on the expressed phenotype, but also on biological pathways in-between (117,118), which aids significantly in functional annotation. Many of the computational techniques behind integrative analysis today rely upon advances in machine learning. The ability of machine learning to make predictions (supervised learning), and find correlations (unsupervised learning), can exceed that of traditional statistical analysis if correctly performed (119).
Most xQTL studies concentrate on correlating sequence variations with trait outcomes. Recent advances in genomic editing technologies, such as Cre-LoxP (120) and CRISPR-Cas9 (121), allow for testing the effects of genomic variations in an experimental setting. Based on xQTL data, a researcher can choose to replicate candidate variants in a model animal, tissue chips or in primary/iPSC cells. An additional approach is to use CRISPR interference (CRISPRi) (122,123), or CRISPR activation (CRISPRa) (124), to induce sequence-specific repression or expression of particular sections of the genome to better our understanding of those gene’s functions (125). CRISPRi and CRISPRa are also ideal for examining how gene expression can affect the development of organs and tissues themselves (126).
At the root of this so-called ‘precision medicine’ is the idea of tailoring treatment to a patient’s individual biology and variants. Recent studies have showed the elevated successful rates of drug target discovery and drug development from genetic and genomic discoveries (127). Artificial intelligence (AI) and network-based methodologies offer a powerful framework to identify risk genes and drug targets from genetic, genomic and xQTL discoveries (128–131). We believe that a genome-wide, integrative AI framework (Fig. 2) to identify novel risk genes and networks from massive genetic and xQTL data will enable a more complete mechanistic understanding of the disease and the development of targeted therapeutic approaches for human complex diseases if broadly applied.
Figure 2.
Diagram illustrating a genome-wide, integrative AI framework to identify risk genes and drug targets from genetic, genomic and xQTL (pQTL, eQTL, sQTL, hQTL and meQTL) findings.
Conflict of Interest statement. The authors have declared no competing interests.
Funding
National Institute of Aging (NIA) of the National Institutes of Health (NIH) (under Award Number U01AG073323, R01AG076448, R01AG066707 and R56AG074001 to F.C.); Sondra J. and Stephen R. Hardis Endowed Chair of Cancer Genomic Medicine at the Cleveland Clinic (to C.E.).
Authors’ contributions
M.B., Y.H., F.C. and C.E. wrote the draft and critically revised the manuscript. All authors critically revised the manuscript and gave final approval.
Contributor Information
Marina Bykova, Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
Yuan Hou, Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
Charis Eng, Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
Feixiong Cheng, Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA.
References
- 1. Dunham, I., Kundaje, A., Aldred, S.F., Collins, P.J., Davis, C.A., Doyle, F., Epstein, C.B., Frietze, S., Harrow, J., Kaul, R. et al. (2012) An integrated encyclopedia of DNA elements in the human genome. Nature, 489, 57–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Haberle, V. and Stark, A. (2018) Eukaryotic core promoters and the functional basis of transcription initiation. Nat. Rev. Mol. Cell Biol., 19, 621–637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Cech, T.R. and Steitz, J.A. (2014) The noncoding RNA revolution-trashing old rules to forge new ones. Cell, 157, 77–94. [DOI] [PubMed] [Google Scholar]
- 4. Jo, B.S. and Choi, S.S. (2015) introns: the functional benefits of introns in genomes. Genomics Inform, 13, 112–118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Misteli, T. (2020) The self-organizing genome: principles of genome architecture and function. Cell, 183, 28–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Zhang, F. and Lupski, J.R. (2015) Non-coding genetic variants in human disease. Hum. Mol. Genet., 24, R102–R110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Wells, A., Heckerman, D., Torkamani, A., Yin, L., Sebat, J., Ren, B., Telenti, A. and di Iulio, J. (2019) Ranking of non-coding pathogenic variants and putative essential regions of the human genome. Nat. Commun., 10, 5241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Esteller, M. (2011) Non-coding RNAs in human disease. Nat. Rev. Genet., 12, 861–874. [DOI] [PubMed] [Google Scholar]
- 9. Brem, R.B., Yvert, G., Clinton, R. and Kruglyak, L. (2002) Genetic dissection of transcriptional regulation in budding yeast. Science, 296, 752–755. [DOI] [PubMed] [Google Scholar]
- 10. Buccitelli, C. and Selbach, M. (2020) mRNAs, proteins and the emerging principles of gene expression control. Nat. Rev. Genet., 21, 630–644. [DOI] [PubMed] [Google Scholar]
- 11. Koussounadis, A., Langdon, S.P., Um, I.H., Harrison, D.J. and Smith, V.A. (2015) Relationship between differentially expressed mRNA and mRNA-protein correlations in a xenograft model system. Sci. Rep., 5, 10775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Nica, A.C. and Dermitzakis, E.T. (2013) Expression quantitative trait loci: present and future. Philos. Trans. R. Soc. Lond. Ser. B Biol. Sci., 368, 20120362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Wright, F.A., Shabalin, A.A. and Rusyn, I. (2012) Computational tools for discovery and interpretation of expression quantitative trait loci. Pharmacogenomics, 13, 343–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Zeng, B., Bendl, J., Kosoy, R., Fullard, J.F., Hoffman, G.E. and Roussos, P. (2022) Multi-ancestry eQTL meta-analysis of human brain identifies candidate causal variants for brain-related traits. Nat. Genet., 54, 161–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Mizuno, A. and Okada, Y. (2019) Biological characterization of expression quantitative trait loci (eQTLs) showing tissue-specific opposite directional effects. Eur. J. Hum. Genet., 27, 1745–1756. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. GTEx Consortium (2020) The GTEx Consortium atlas of genetic regulatory effects across human tissues. Science, 369, 1318–1330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Barbeira, A.N., Bonazzola, R., Gamazon, E.R., Liang, Y., Park, Y., Kim-Hellmuth, S., Wang, G., Jiang, Z., Zhou, D., Hormozdiari, F. et al. (2021) Exploiting the GTEx resources to decipher the mechanisms at GWAS loci. Genome Biol., 22, 49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Davenport, E.E., Amariuta, T., Gutierrez-Arcelus, M., Slowikowski, K., Westra, H.-J., Luo, Y., Shen, C., Rao, D.A., Zhang, Y., Pearson, S. et al. (2018) Discovering in vivo cytokine-eQTL interactions from a lupus clinical trial. Genome Biol., 19, 168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Rong, Y., Dong, S.S., Hu, W.X., Guo, Y., Chen, Y.X., Chen, J.B., Zhu, D.L., Chen, H., Yang, T.L. (2021) Ddrs: Detection of drug response snps specifically in patients receiving drug treatment. Comput Struct Biotechnol J, 19, 3650–3657. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Nie, L., Wu, G. and Zhang, W. (2006) Correlation of mRNA expression and protein abundance affected by multiple sequence features related to translational efficiency in Desulfovibrio vulgaris: a quantitative analysis. Genetics, 174, 2229–2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Xie, F., Liu, T., Qian, W.J., Petyuk, V.A. and Smith, R.D. (2011) Liquid chromatography-mass spectrometry-based quantitative proteomics. J. Biol. Chem., 286, 25443–25449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Schwanhäusser, B., Busse, D., Li, N., Dittmar, G., Schuchhardt, J., Wolf, J., Chen, W. and Selbach, M. (2011) Global quantification of mammalian gene expression control. Nature, 473, 337–342. [DOI] [PubMed] [Google Scholar]
- 23. Richards, A.L., Eckhardt, M. and Krogan, N.J. (2021) Mass spectrometry-based protein-protein interaction networks for the study of human diseases. Mol. Syst. Biol., 17, e8792. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Yang, C., Farias, F.H.G., Ibanez, L., Suhy, A., Sadler, B., Fernandez, M.V., Wang, F., Bradley, J.L., Eiffert, B., Bahena, J.A. et al. (2021) Genomic atlas of the proteome from brain, CSF and plasma prioritizes proteins implicated in neurological disorders. Nat. Neurosci., 24, 1302–1312. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Yao, C., Chen, G., Song, C., Keefe, J., Mendelson, M., Huan, T., Sun, B.B., Laser, A., Maranville, J.C., Wu, H. et al. (2018) Genome-wide mapping of plasma protein QTLs identifies putatively causal genes and pathways for cardiovascular disease. Nat. Commun., 9, 3268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Zhang, J., Dutta, D., Kottgen, A., Tin, A., Schlosser, P., Grams, M.E., Harvey, B., Consortium, C.K., Yu, B., Boerwinkle, E. et al. (2022) Plasma proteome analyses in individuals of european and african ancestry identify cis-pqtls and models for proteome-wide association studies. Nat Genet, 54, 593–602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Workman, J.L. and Kingston, R.E. (1998) Alteration of nucleosome structure as a mechanism of transcriptional regulation. Annu. Rev. Biochem., 67, 545–579. [DOI] [PubMed] [Google Scholar]
- 28. Korolev, N., Vorontsova, O.V. and Nordenskiöld, L. (2007) Physicochemical analysis of electrostatic foundation for DNA-protein interactions in chromatin transformations. Prog. Biophys. Mol. Biol., 95, 23–49. [DOI] [PubMed] [Google Scholar]
- 29. Bannister, A.J. and Kouzarides, T. (2011) Regulation of chromatin by histone modifications. Cell Res., 21, 381–395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Zhang, L., Xue, G., Liu, J., Li, Q. and Wang, Y. (2018) Revealing transcription factor and histone modification co-localization and dynamics across cell lines by integrating ChIP-seq and RNA-seq data. BMC Genomics, 19, 914. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Schmidt, F., Gasparoni, N., Gasparoni, G., Gianmoena, K., Cadenas, C., Polansky, J.K., Ebert, P., Nordström, K., Barann, M., Sinha, A. et al. (2017) Combining transcription factor binding affinities with open-chromatin data for accurate gene expression prediction. Nucleic Acids Res., 45, 54–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Yi, S.J. and Kim, K. (2020) New insights into the role of histone changes in aging. Int. J. Mol. Sci., 21, 8241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Fang, L., Wuptra, K., Chen, D., Li, H., Huang, S.K., Jin, C. and Yokoyama, K.K. (2014) Environmental-stress-induced chromatin regulation and its heritability. J Carcinog Mutagen, 5, 1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Audia, J.E. and Campbell, R.M. (2016) Histone modifications and cancer. Cold Spring Harb. Perspect. Biol., 8, a019521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Du, J., Johnson, L.M., Jacobsen, S.E. and Patel, D.J. (2015) DNA methylation pathways and their crosstalk with histone methylation. Nat. Rev. Mol. Cell Biol., 16, 519–532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Rose, N.R. and Klose, R.J. (2014) Understanding the relationship between DNA methylation and histone lysine methylation. Biochim. Biophys. Acta, 1839, 1362–1372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. McVicker, G., van de Geijn, B., Degner, J.F., Cain, C.E., Banovich, N.E., Raj, A., Lewellen, N., Myrthil, M., Gilad, Y. and Pritchard, J.K. (2013) Identification of genetic variants that affect histone modifications in human cells. Science, 342, 747–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Pelikan, R.C., Kelly, J.A., Fu, Y., Lareau, C.A., Tessneer, K.L., Wiley, G.B., Wiley, M.M., Glenn, S.B., Harley, J.B., Guthridge, J.M. et al. (2018) Enhancer histone-QTLs are enriched on autoimmune risk haplotypes and influence gene expression within chromatin networks. Nat. Commun., 9, 2905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Grubert, F., Zaugg, J.B., Kasowski, M., Ursu, O., Spacek, D.V., Martin, A.R., Greenside, P., Srivas, R., Phanstiel, D.H., Pekowska, A. et al. (2015) Genetic control of chromatin states in humans involves local and distal chromosomal interactions. Cell, 162, 1051–1065. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Wang, E.T., Sandberg, R., Luo, S., Khrebtukova, I., Zhang, L., Mayr, C., Kingsmore, S.F., Schroth, G.P. and Burge, C.B. (2008) Alternative isoform regulation in human tissue transcriptomes. Nature, 456, 470–476. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Noh, S.-J., Lee, K., Paik, H. and Hur, C.-G. (2006) TISA: Tissue-specific alternative splicing in human and mouse genes. DNA Res., 13, 229–243. [DOI] [PubMed] [Google Scholar]
- 42. Tian, G.G., Li, J. and Wu, J. (2020) Alternative splicing signatures in preimplantation embryo development. Cell Biosci., 10, 33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Weyn-Vanhentenryck, S.M., Feng, H., Ustianenko, D., Duffié, R., Yan, Q., Jacko, M., Martinez, J.C., Goodwin, M., Zhang, X., Hengst, U. et al. (2018) Precise temporal regulation of alternative splicing during neural development. Nat. Commun., 9, 2189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Love, J.E., Hayden, E.J. and Rohn, T.T. (2015) Alternative splicing in Alzheimer's disease. J Parkinsons Dis Alzheimers Dis, 2, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Cherry, J.D., Esnault, C.D., Baucom, Z.H., Tripodis, Y., Huber, B.R., Alvarez, V.E., Stein, T.D., Dickson, D.W. and McKee, A.C. (2021) Tau isoforms are differentially expressed across the hippocampus in chronic traumatic encephalopathy and Alzheimer’s disease. Acta Neuropathol. Commun., 9, 86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. Griffin, C. and Saint-Jeannet, J.P. (2020) Spliceosomopathies: diseases and mechanisms. Dev. Dyn., 249, 1038–1046. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Bai, B., Hales, C.M., Chen, P.-C., Gozal, Y., Dammer, E.B., Fritz, J.J., Wang, X., Xia, Q., Duong, D.M., Street, C. et al. (2013) U1 small nuclear ribonucleoprotein complex and RNA splicing alterations in Alzheimer’s disease. Proc. Natl. Acad. Sci., 110, 16562–16567. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Raj, T., Li, Y.I., Wong, G., Humphrey, J., Wang, M., Ramdhani, S., Wang, Y.C., Ng, B., Gupta, I., Haroutunian, V. et al. (2018) Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer's disease susceptibility. Nat. Genet., 50, 1584–1592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Zhang, Y., Yang, H.T., Kadash-Edmondson, K., Pan, Y., Pan, Z., Davidson, B.L. and Xing, Y. (2020) Regional variation of splicing QTLs in human brain. Am. J. Hum. Genet., 107, 196–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Walker, R.L., Ramaswami, G., Hartl, C., Mancuso, N., Gandal, M.J., de la Torre-Ubieta, L., Pasaniuc, B., Stein, J.L. and Geschwind, D.H. (2019) Genetic control of expression and splicing in developing human brain informs disease mechanisms. Cell, 179, 750–771.e722. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Nishiyama, A. and Nakanishi, M. (2021) Navigating the DNA methylation landscape of cancer. Trends Genet., 37, 1012–1027. [DOI] [PubMed] [Google Scholar]
- 52. Locke, W.J., Guanzon, D., Ma, C., Liew, Y.J., Duesing, K.R., Fung, K.Y.C. and Ross, J.P. (2019) DNA methylation cancer biomarkers: translation to the clinic. Front. Genet., 10, 1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Clark, A.D., Nair, N., Anderson, A.E., Thalayasingam, N., Naamane, N., Skelton, A.J., Diboll, J., Barton, A., Eyre, S., Isaacs, J.D. et al. (2020) Lymphocyte DNA methylation mediates genetic risk at shared immune-mediated disease loci. J. Allergy Clin. Immunol., 145, 1438–1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Liu, D., Wang, Y., Jing, H., Meng, Q. and Yang, J. (2022) Novel DNA methylation loci and genes showing pleiotropic association with Alzheimer's dementia: a network Mendelian randomization analysis. Epigenetics, 17, 746–758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Banovich, N.E., Lan, X., McVicker, G., van de Geijn, B., Degner, J.F., Blischak, J.D., Roux, J., Pritchard, J.K. and Gilad, Y. (2014) Methylation QTLs are associated with coordinated changes in transcription factor binding, histone modifications, and gene expression levels. PLoS Genet., 10, e1004663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Hop, P.J., Luijk, R., Daxinger, L., van Iterson, M., Dekkers, K.F., Jansen, R., Heijmans, B.T., t Hoen, P.A.C., van Meurs, J., Jansen, R. et al. (2020) Genome-wide identification of genes regulating DNA methylation using genetic anchors for causal inference. Genome Biol., 21, 220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Junot, C., Fenaille, F., Colsch, B. and Bécher, F. (2014) High resolution mass spectrometry based techniques at the crossroads of metabolic pathways. Mass Spectrom. Rev., 33, 471–500. [DOI] [PubMed] [Google Scholar]
- 58. Yin, X., Chan, L.S., Bose, D., Jackson, A.U., VandeHaar, P., Locke, A.E., Fuchsberger, C., Stringham, H.M., Welch, R., Yu, K. et al. (2022) Genome-wide association studies of metabolites in Finnish men identify disease-relevant loci. Nat. Commun., 13, 1644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Nicholson, G., Rantalainen, M., Li, J.V., Maher, A.D., Malmodin, D., Ahmadi, K.R., Faber, J.H., Barrett, A., Min, J.L., Rayner, N.W. et al. (2011) A genome-wide metabolic QTL analysis in europeans implicates two loci shaped by recent positive selection. PLoS Genet., 7, e1002270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Tang, Z.-Z., Chen, G., Hong, Q., Huang, S., Smith, H.M., Shah, R.D., Scholz, M. and Ferguson, J.F. (2019) Multi-omic analysis of the microbiome and metabolome in healthy subjects reveals microbiome-dependent relationships between diet and metabolites. Front. Genet., 10, 454. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Belheouane, M., Gupta, Y., Künzel, S., Ibrahim, S. and Baines, J.F. (2017) Improved detection of gene–microbe interactions in the mouse skin microbiota using high-resolution QTL mapping of 16S rRNA transcripts. Microbiome, 5, 59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Gupta, V.K., Kim, M., Bakshi, U., Cunningham, K.Y., Davis, J.M., Lazaridis, K.N., Nelson, H., Chia, N. and Sung, J. (2020) A predictive index for health status using species-level gut microbiome profiling. Nat. Commun., 11, 4635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Bycroft, C., Freeman, C., Petkova, D., Band, G., Elliott, L.T., Sharp, K., Motyer, A., Vukcevic, D., Delaneau, O., O’Connell, J. et al. (2018) The uk biobank resource with deep phenotyping and genomic data. Nature, 562, 203–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Sudlow, C., Gallacher, J., Allen, N., Beral, V., Burton, P., Danesh, J., Downey, P., Elliott, P., Green, J., Landray, M. et al. (2015) UK Biobank: an open access resource for identifying the causes of a wide range of complex diseases of middle and old age. PLoS Med., 12, e1001779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Taliun, D., Harris, D.N., Kessler, M.D., Carlson, J., Szpiech, Z.A., Torres, R., Taliun, S.A.G., Corvelo, A., Gogarten, S.M., Kang, H.M. et al. (2021) Sequencing of 53,831 diverse genomes from the NHLBI TOPMed Program. Nature, 590, 290–299. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. The All of Us Research Program Investigators (2019) The “All of Us” Research Program. N. Engl. J. Med., 381, 668–676. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Kanai, M., Akiyama, M., Takahashi, A., Matoba, N., Momozawa, Y., Ikeda, M., Iwata, N., Ikegawa, S., Hirata, M., Matsuda, K. et al. (2018) Genetic analysis of quantitative traits in the japanese population links cell types to complex human diseases. Nat Genet, 50, 390–400. [DOI] [PubMed] [Google Scholar]
- 68. Nagai, A., Hirata, M., Kamatani, Y., Muto, K., Matsuda, K., Kiyohara, Y., Ninomiya, T., Tamakoshi, A., Yamagata, Z., Mushiroda, T. et al. (2017) Overview of the BioBank Japan Project: study design and profile. J. Epidemiol., 27, S2–s8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Buniello, A., MacArthur, J.A.L., Cerezo, M., Harris, L.W., Hayhurst, J., Malangone, C., McMahon, A., Morales, J., Mountjoy, E., Sollis, E. et al. (2018) The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res., 47, D1005–D1012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Halldorsson, B.V., Eggertsson, H.P., Moore, K.H.S., Hauswedell, H., Eiriksson, O., Ulfarsson, M.O., Palsson, G., Hardarson, M.T., Oddsson, A., Jensson, B.O. et al. (2022) The sequences of 150,119 genomes in the UK Biobank. Nature, 607, 732–740. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Devaney, S. All of US . (2019) Nature, 576, S15–S17. [Google Scholar]
- 72. Mapes, B.M., Foster, C.S., Kusnoor, S.V., Epelbaum, M.I., AuYoung, M., Jenkins, G., Lopez-Class, M., Richardson-Heron, D., Elmi, A., Surkan, K. et al. (2020) Diversity and inclusion for the All of Us research program: a scoping review. PLoS One, 15, e0234962. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Beecham, G.W., Bis, J.C., Martin, E.R., Choi, S.H., DeStefano, A.L., van Duijn, C.M., Fornage, M., Gabriel, S.B., Koboldt, D.C., Larson, D.E. et al. (2017) The Alzheimer's Disease Sequencing Project: study design and sample selection. Neurol. Genet., 3, e194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Petersen, R.C., Aisen, P.S., Beckett, L.A., Donohue, M.C., Gamst, A.C., Harvey, D.J., Jack, C.R., Jr., Jagust, W.J., Shaw, L.M., Toga, A.W. et al. (2010) Alzheimer's Disease Neuroimaging Initiative (ADNI): clinical characterization. Neurology, 74, 201–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Kuzma, A., Valladares, O., Cweibel, R., Greenfest-Allen, E., Childress, D.M., Malamon, J., Gangadharan, P., Zhao, Y., Qu, L., Leung, Y.Y. et al. (2016) NIAGADS: the NIA Genetics of Alzheimer's Disease Data Storage Site. Alzheimers Dement., 12, 1200–1203. [Google Scholar]
- 76. Greenwood, A.K., Montgomery, K.S., Kauer, N., Woo, K.H., Leanza, Z.J., Poehlman, W.L., Gockley, J., Sieberts, S.K., Bradic, L., Logsdon, B.A. et al. (2020) The AD knowledge portal: a repository for multi-omic data on Alzheimer's disease and aging. Curr. Protoc. Hum. Genet., 108, e105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Zhou, Y., Fang, J., Bekris, L.M., Kim, Y.H., Pieper, A.A., Leverenz, J.B., Cummings, J. and Cheng, F. (2021) AlzGPS: a genome-wide positioning systems platform to catalyze multi-omics for Alzheimer's drug discovery. Alzheimers Res. Ther., 13, 24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Zhou, Y., Xu, J., Hou, Y., Bekris, Y., Leverenz, J.B., Pieper, A.A., Cummings, J., and Cheng, F. (2022) The Alzheimer’s Cell Atlas (TACA): A single-cell molecular map for translational therapeutics accelerator in Alzheimer’s disease. Alzheimer and Dementia, in press. 10.1002/trc2.12350. [DOI] [PMC free article] [PubMed]
- 79. Abascal, F., Acosta, R., Addleman, N.J., Adrian, J., Afzal, V., Ai, R., Aken, B., Akiyama, J.A., Jammal, O.A., Amrhein, H. et al. (2020) Expanded encyclopaedias of DNA elements in the human and mouse genomes. Nature, 583, 699–710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. ENCODE Project Consortium . (2012) An integrated encyclopedia of DNA elements in the human genome. Nature, 489, 57–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. The GTEx Consortium . (2013) The genotype-tissue expression (gtex) project. Nat Genet, 45, 580–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Andersson, R., Gebhard, C., Miguel-Escalada, I., Hoof, I., Bornholdt, J., Boyd, M., Chen, Y., Zhao, X., Schmidl, C., Suzuki, T. et al. (2014) An atlas of active enhancers across human cell types and tissues. Nature, 507, 455–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Lizio, M., Harshbarger, J., Shimoji, H., Severin, J., Kasukawa, T., Sahin, S., Abugessaisa, I., Fukuda, S., Hori, F., Ishikawa-Kato, S. et al. (2015) Gateways to the FANTOM5 promoter level mammalian expression atlas. Genome Biol., 16, 22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Noguchi, S., Arakawa, T., Fukuda, S., Furuno, M., Hasegawa, A., Hori, F., Ishikawa-Kato, S., Kaida, K., Kaiho, A., Kanamori-Katayama, M. et al. (2017) FANTOM5 CAGE profiles of human and mouse samples. Scientific Data, 4, 170112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Shabalina, S.A. and Spiridonov, N.A. (2004) The mammalian transcriptome and the function of non-coding DNA sequences. Genome Biol., 5, 105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Engreitz, J.M., Haines, J.E., Perez, E.M., Munson, G., Chen, J., Kane, M., McDonel, P.E., Guttman, M. and Lander, E.S. (2016) Local regulation of gene expression by lncRNA promoters, transcription and splicing. Nature, 539, 452–455. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Yao, R.W., Wang, Y. and Chen, L.L. (2019) Cellular functions of long noncoding RNAs. Nat. Cell Biol., 21, 542–551. [DOI] [PubMed] [Google Scholar]
- 88. Yan, Q., Zhu, C., Guang, S. and Feng, X. (2019) The functions of non-coding RNAs in rRNA regulation. Front. Genet., 10, 290. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Takemata, N. and Ohta, K. (2017) Role of non-coding RNA transcription around gene regulatory elements in transcription factor recruitment. RNA Biol., 14, 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Wittkopp, P.J. and Kalay, G. (2012) Cis-regulatory elements: molecular mechanisms and evolutionary processes underlying divergence. Nat. Rev. Genet., 13, 59–69. [DOI] [PubMed] [Google Scholar]
- 91. Espinar, L., Schikora Tamarit, M.A., Domingo, J., Carey, L.B. (2018) Promoter architecture determines cotranslational regulation of mrna. Genome Res, 28, 509–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Quénet, D. and Dalal, Y. (2014) A long non-coding RNA is required for targeting centromeric protein A to the human centromere. elife, 3, e03254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Giral, H., Landmesser, U. and Kratzer, A. (2018) Into the wild: GWAS exploration of non-coding RNAs. Front. Cardiovasc. Med., 5, 181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Tahami Monfared, A.A., Byrnes, M.J., White, L.A. and Zhang, Q. (2022) Alzheimer’s disease: epidemiology and clinical progression. Neurol. Therapy, 11, 553–569. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Bekris, L.M., Yu, C.E., Bird, T.D. and Tsuang, D.W. (2010) Genetics of Alzheimer disease. J. Geriatr. Psychiatry Neurol., 23, 213–227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Saunders, A.M., Strittmatter, W.J., Schmechel, D., George-Hyslop, P.H., Pericak-Vance, M.A., Joo, S.H., Rosi, B.L., Gusella, J.F., Crapper-MacLachlan, D.R., Alberts, M.J. et al. (1993) Association of apolipoprotein E allele epsilon 4 with late-onset familial and sporadic Alzheimer's disease. Neurology, 43, 1467–1472. [DOI] [PubMed] [Google Scholar]
- 97. Blue, E.E., Cheng, A., Chen, S. and Yu, C.E. (2020) Association of uncommon, noncoding variants in the APOE region with risk of Alzheimer disease in adults of European ancestry. JAMA Netw. Open, 3, e2017666. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Zhou, X., Chen, Y., Mok, K.Y., Kwok, T.C.Y., Mok, V.C.T., Guo, Q., Ip, F.C., Chen, Y., Mullapudi, N., Giusti-Rodríguez, P. et al. (2019) Non-coding variability at the APOE locus contributes to the Alzheimer's risk. Nat. Commun., 10, 3310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Amlie-Wolf, A., Tang, M., Way, J., Dombroski, B., Jiang, M., Vrettos, N., Chou, Y.-F., Zhao, Y., Kuzma, A., Mlynarski, E.E. et al. (2019) Inferring the molecular mechanisms of noncoding Alzheimer's disease-associated genetic variants. J. Alzheimers Dis., 72, 301–318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Kunkle, B.W., Grenier-Boley, B., Sims, R., Bis, J.C., Damotte, V., Naj, A.C., Boland, A., Vronskaya, M., van der Lee, S.J., Amlie-Wolf, A. et al. (2019) Genetic meta-analysis of diagnosed Alzheimer’s disease identifies new risk loci and implicates Aβ, tau, immunity and lipid processing. Nat. Genet., 51, 414–430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Hollingworth, P., Harold, D., Sims, R., Gerrish, A., Lambert, J.C., Carrasquillo, M.M., Abraham, R., Hamshere, M.L., Pahwa, J.S., Moskvina, V. et al. (2011) Common variants at ABCA7, MS4A6A/MS4A4E, EPHA1, CD33 and CD2AP are associated with Alzheimer's disease. Nat. Genet., 43, 429–435. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102. Li, Y.I., Knowles, D.A., Humphrey, J., Barbeira, A.N., Dickinson, S.P., Im, H.K. and Pritchard, J.K. (2018) Annotation-free quantification of RNA splicing using LeafCutter. Nat. Genet., 50, 151–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Zhao, T., Hu, Y., Zang, T. and Wang, Y. (2019) Integrate GWAS, eQTL, and mQTL data to identify Alzheimer’s disease-related genes. Front. Genet., 10, 1021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Wang, Q., Chen, Y., Readhead, B., Chen, K., Su, Y., Reiman, E.M. and Dudley, J.T. (2020) Longitudinal data in peripheral blood confirm that PM20D1 is a quantitative trait locus (QTL) for Alzheimer’s disease and implicate its dynamic role in disease progression. Clin. Epigenetics, 12, 189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Hillary, R.F., Gadd, D.A., McCartney, D.L., Shi, L., Campbell, A., Walker, R.M., Ritchie, C.W., Deary, I.J., Evans, K.L., Nevado-Holgado, A.J. et al. (2022) Genome- and epigenome-wide studies of plasma protein biomarkers for Alzheimer's disease implicate TBCA and TREM2 in disease risk. Alzheimers Dement (Amst), 14, e12280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Ng, B., White, C.C., Klein, H.U., Sieberts, S.K., McCabe, C., Patrick, E., Xu, J., Yu, L., Gaiteri, C., Bennett, D.A. et al. (2017) An xQTL map integrates the genetic architecture of the human brain's transcriptome and epigenome. Nat. Neurosci., 20, 1418–1426. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Novikova, G., Kapoor, M., Tcw, J., Abud, E.M., Efthymiou, A.G., Chen, S.X., Cheng, H., Fullard, J.F., Bendl, J., Liu, Y. et al. (2021) Integration of Alzheimer’s disease genetics and myeloid genomics identifies disease risk regulatory elements and genes. Nat. Commun., 12, 1610. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Goswami, C., Chattopadhyay, A. and Chuang, E.Y. (2021) Rare variants: data types and analysis strategies. Ann. Transl. Med., 9, 961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Luedtke, A., Powers, S., Petersen, A., Sitarik, A., Bekmetjev, A. and Tintle, N.L. (2011) Evaluating methods for the analysis of rare variants in sequence data. BMC Proc., 5, S119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Curk, T., Rot, G. and Zupan, B. (2011) SNPsyn: detection and exploration of SNP-SNP interactions. Nucleic Acids Res., 39, W444–W449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Hahn, L.W., Ritchie, M.D. and Moore, J.H. (2003) Multifactor dimensionality reduction software for detecting gene–gene and gene–environment interactions. Bioinformatics, 19, 376–382. [DOI] [PubMed] [Google Scholar]
- 112. Choudhury, A., Hazelhurst, S., Meintjes, A., Achinike-Oduaran, O., Aron, S., Gamieldien, J., Jalali Sefid Dashti, M., Mulder, N., Tiffin, N. and Ramsay, M. (2014) Population-specific common SNPs reflect demographic histories and highlight regions of genomic plasticity with functional relevance. BMC Genomics, 15, 437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Hellwege, J.N., Keaton, J.M., Giri, A., Gao, X., Velez Edwards, D.R. and Edwards, T.L. (2017) population stratification in genetic association studies. Curr. Protoc. Hum. Genet., 95, 1.22.21-21.22.23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Mills, M.C. and Rahal, C. (2019) A scientometric review of genome-wide association studies. Commun. Biol., 2, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Sirugo, G., Williams, S.M. and Tishkoff, S.A. (2019) The missing diversity in human genetic studies. Cell, 177, 26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Mulvey, B., Lagunas, T., Jr., Dougherty, J.D. (2021) Massively parallel reporter assays: Defining functional psychiatric genetic variants across biological contexts. Biol Psychiatry, 89, 76–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Keele, G.R., Quach, B.C., Israel, J.W., Chappell, G.A., Lewis, L., Safi, A., Simon, J.M., Cotney, P., Crawford, G.E., Valdar, W. et al. (2020) Integrative qtl analysis of gene expression and chromatin accessibility identifies multi-tissue patterns of genetic regulation. PLoS Genet, 16, e1008537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Taylor, D.L., Jackson, A.U., Narisu, N., Hemani, G., Erdos, M.R., Chines, P.S., Swift, A., Idol, J., Didion, J.P., Welch, R.P. et al. (2019) Integrative analysis of gene expression, DNA methylation, physiological traits, and genetic variation in human skeletal muscle. Proc. Natl. Acad. Sci., 116, 10883–10888. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Nicholls, H.L., John, C.R., Watson, D.S., Munroe, P.B., Barnes, M.R. and Cabrera, C.P. (2020) Reaching the end-game for GWAS: machine learning approaches for the prioritization of complex disease loci. Front. Genet., 11, 350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Kim, H., Kim, M., Im, S.K. and Fang, S. (2018) Mouse Cre-LoxP system: general principles to determine tissue-specific roles of target genes. Lab. Anim. Res., 34, 147–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Uddin, F., Rudin, C.M. and Sen, T. (2020) CRISPR gene therapy: applications, limitations, and implications for the future. Front. Oncol., 10, 1387. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Larson, M.H., Gilbert, L.A., Wang, X., Lim, W.A., Weissman, J.S. and Qi, L.S. (2013) CRISPR interference (CRISPRi) for sequence-specific control of gene expression. Nat. Protoc., 8, 2180–2196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Duke, C.G., Bach, S.V., Revanna, J.S., Sultan, F.A., Southern, N.T., Davis, M.N., Carullo, N.V.N., Bauman, A.J., Phillips, R.A., 3rd and Day, J.J. (2020) An improved CRISPR/dCas9 interference tool for neuronal gene suppression. Front. Genome Ed., 2, 9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Chen, M. and Qi, L.S. (2017) Repurposing CRISPR system for transcriptional activation. Adv. Exp. Med. Biol., 983, 147–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Ren, X., Wang, M., Li, B., Jamieson, K., Zheng, L., Jones, I.R., Li, B., Takagi, M.A., Lee, J., Maliskova, L. et al. (2021) Parallel characterization of cis-regulatory elements for multiple genes using CRISPRpath. Sci. Adv., 7, eabi4360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Song, M., Pebworth, M.-P., Yang, X., Abnousi, A., Fan, C., Wen, J., Rosen, J.D., Choudhary, M.N.K., Cui, X., Jones, I.R. et al. (2020) Cell-type-specific 3D epigenomes in the developing human cortex. Nature, 587, 644–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Reay, W.R. and Cairns, M.J. (2021) Advancing the use of genome-wide association studies for drug repurposing. Nat. Rev. Genet., 22, 658–671. [DOI] [PubMed] [Google Scholar]
- 128. Wang, Q., Chen, R., Cheng, F., Wei, Q., Ji, Y., Yang, H., Zhong, X., Tao, R., Wen, Z., Sutcliffe, J.S. et al. (2019) A Bayesian framework that integrates multi-omics data and gene networks predicts risk genes from schizophrenia GWAS data. Nat. Neurosci., 22, 691–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Fang, J., Zhang, P., Wang, Q., Chiang, C.W., Zhou, Y., Hou, Y., Xu, J., Chen, R., Zhang, B., Lewis, S.J. et al. (2022) Artificial intelligence framework identifies candidate targets for drug repurposing in Alzheimer's disease. Alzheimers Res. Ther., 14, 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Fang, J., Zhang, P., Zhou, Y., Chiang, C.W., Tan, J., Stauffer, S., Li, L., Pieper, A., Cummings, J. and Cheng, F. (2021) Endophenotype-based in-silico network medicine discovery combined with insurance records data mining identifies sildenafil as a candidate drug for Alzheimer’s disease. Nat. Aging, 1, 1175–1188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Xu, J., Zhang, P., Huang, Y., Zhou, Y., Hou, Y., Bekris, L., Lathia, J., Chiang, C.W., Li, L., Pieper, A. et al. (2021) Multimodal single-cell/nucleus RNA sequencing data analysis uncovers molecular networks between disease-associated microglia and astrocytes with implications for drug repurposing in Alzheimer's disease. Genome Res., 31, 1900–1912. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Greenwood, A.K., Montgomery, K.S., Kauer, N., Woo, K.H., Leanza, Z.J., Poehlman, W.L., Gockley, J., Sieberts, S.K., Bradic, L., Logsdon, B.A. et al. (2020) The ad knowledge portal: A repository for multi-omic data on alzheimer’s disease and aging. Curr Protoc Hum Genet, 108, e105. [DOI] [PMC free article] [PubMed] [Google Scholar]