Abstract
Genome-Wide Association Studies (GWAS) have elucidated the genetic components of Parkinson's Disease (PD). However, the vast majority of GWAS association signals fall within non-coding regions, translating these results into an interpretable, mechanistic understanding of the disease etiology remains a major challenge in the field. In this review, we provide an overview of the approaches to prioritize putative causal variants and genes as well as summarise the primary findings of previous studies. We then discuss recent progress made in efforts to integrate multi-omics data to identify likely pathogenic cell types and biological pathways implicated in PD pathogenesis. We have compiled complete PD GWAS cell-type and tissue enrichment summary statistics from multiple studies and provided them in a standardized format as a resource for the research community to use in meta-analyses. Additionally, we discuss experimental approaches that will be needed to further elucidate the effects of functional variants and genes on cellular phenotypes and disease risk.
Keywords: Parkinson’s Disease, Neurodegeneration, Genome-Wide Association Study (GWAS), Quantitative Trait Loci (QTL), Multi-omics
Introduction
Parkinson's disease (PD) (Parkinson, 1817) is the second most common neurodegenerative disease globally (Kim et al., 2018) and is projected to double in prevalence between 2005 and 2030 (Dorsey et al., 2007). PD is characterized by both motor symptoms (e.g. bradykinesia, rigidity, tremor) and a diversity of non-motor symptoms (e.g. cognitive impairment, autonomic dysfunction, sleep cycle dysregulation), but there is substantial heterogeneity in the exact clinical presentation and age of onset, as well as phenotypic overlap with other PD-like disorders (Poewe et al., 2017). This phenotypic heterogeneity, which is potentially a reflection of heterogeneity at the molecular level, has further compounded the challenge of developing methodologies that can effectively identify, treat and prevent this disorder.
PD is characterized by two main pathological hallmarks: the loss of dopaminergic (DA) neurons in the substantia nigra (SN) pars compacta (Damier et al., 1999; Fearnley and Lees, 1991) and the accumulation of Lewy bodies, caused by the intracellular aggregation of α-synuclein (Braak et al., 2003; Braak and Del Tredici, 2017; Poewe et al., 2017). This leads to the dysfunction of basal ganglia circuitry and is thought to be the primary driver of classical motor symptoms (Kalia and Lang, 2015). The main pharmacological intervention is levodopa replacement but there is currently no cure for the disease. Normally PD cases are categorized in two main forms: monogenic and idiopathic. The so-called monogenic cases (sometimes called Mendelian, familial, or genetic) only account for 5-10% of total cases (Sara Bandres-Ciga et al., 2020; Lesage and Brice, 2009). The vast majority of PD cases are idiopathic (also called sporadic or sometimes “non-genetic”), which we now know has a substantial genetic component that comprises many common, small-effect variants across the genome. PD has been estimated to be anywhere from 34% heritable based on twin studies (Wirdefeldt et al., 2011), to 22-27% heritable based on genetic (SNP-based) studies (Blauwendraat et al., 2020; Chang et al., 2017; Keller et al., 2012). Despite large-scale multi-national efforts, at present known genetic factors can only explain 22-36% of PD risk heritability in populations of European descent (Nalls et al., 2019a). Even the monogenic forms of PD do not have 100% penetrance and therefore are likely modified by genetic background as well as environmental interactions. As for this review, we will focus specifically on the idiopathic form of PD.
Technological advances are allowing the surge of multi-omics which are accelerating our understanding of this disease. In this review, our objective is not necessarily to enumerate all variants and genes that have been implicated in PD pathology (for these details, see: (Sara Bandres-Ciga et al., 2020; Blauwendraat et al., 2020; Nalls et al., 2019a; Redenšek et al., 2018)). Instead, we will highlight methods of investigation across multiple scales of biology (variants, genes, pathways, cell-types, tissues, phenotypes), direct readers to studies, tools, and datasets relevant to PD -omics (Table 1), bring attention to conceptual challenges in the field while suggesting some potential avenues of progress. We have also compiled and standardised cell-type and tissue enrichment results (Table S1) and correlations between PD and other phenotypes (Table S2) across multiple studies of PD genomics. Through this, we hope to promote a more cohesive understanding of the complex biology of PD in the service of ultimately improving the lives of those afflicted by this disease.
Table 1.
Resource | Phenotypes | Description | Data types |
Levels | Last update |
No request needed |
API | URL | Reference |
---|---|---|---|---|---|---|---|---|---|
Alfradique-Dunham et al. (2021) | Parkinson's | Full genome-wide summary statistics from Alfradique-Dunham et al. (2021) PD motor subtypes GWAS. | GWAS | Variants | 2021 | Y | N | https://pdgenetics.org/resources | (Alfradique-Dunham et al. 2021) |
AMP-PD | Parkinson's | Whole-genome sequencing (WGS), familial PD-associated mutation status, transcriptome and clinical evaluations in a cohort of PD and control participants. Some of the data is compiled from other databases (BioFIND, PDBP, PPMI) and harmonized to facilitate re-analysis. |
WGS, bulkRNA-seq | Variants, Genes, Phenotypes | 2021 | N | Y | https://amp-pd.org | (Iwaki et al. 2020) |
Blauwendraat et al. (2019) | Parkinson's | Full genome-wide summary statistics from Blauwendraat et al. (2019) PD onset GWAS, excluding 23andMe. | GWAS | Variants | 2019 | Y | N | https://pdgenetics.org/resources | (Blauwendraat et al. 2019) |
FOUNDIN-PD | Parkinson's | Extensive multi-omics resource from PD patient-derived iPSCs. | Genotype, WGS, mitoDNA-seq DNA methylation, HiC, scATAC-seq, bulkRNA-seq, smallRNA-seq, scRNA-seq, Proteomics, Clinical, Imaging | Variants, Epigenomics, Genes, Cell-types, Tissues, Phenotypes | 2021 | Y | Y | https://www.foundinpd.org | (Bressan et al., 2021) |
GP2 | Parkinson's | The Global Parkinson’s Genetics Program (GP2) is the first resource project of the Aligning Science Across Parkinson's (ASAP) initiative. It aims to collect multi-modal data from a diverse international cohort of PD patients, including genotyping in 150k+ individuals and WGS in 10k+ individuals. | Genotype, GWAS, WGS | Variants, Epigenomics, Genes, Cell-types, Tissues, Phenotypes | 2021 | N | Y | https://www.gp2.org | (The Global Parkinson's Genetics Program, 2021) |
iPDGC Locus Browser | Parkinson's | Top summary statistics from three PD GWAS with variant annotation, causal gene(s) nomination, brain eQTL integration, statistical fine-mapping results, co-expression networks and burden analyses. | GWAS, QTL, bulkRNA-seq, scRNA-seq | Variants, Genes, Phenotypes | 2021 | Y | N | https://pdgenetics.shinyapps.io/GWASBrowser | (Grenn et al. 2020) |
iPDGC MR Portal | Parkinson's | Two-sample Mendelian Randomisation (MR) comparing PD to 5,854 other GWAS traits provided by the International Parkinson Disease Genomics Consortium (IPDGC). | GWAS | Variants, Phenotypes | 2019 | Y | N | https://pdgenetics.shinyapps.io/MRportal | (Noyce et al. 2019) |
MJFF | Parkinson's | A compilation of PD genomics datasets and tools funded by MIchael J. Fox Foundation (MJFF). | GWAS, WGS, bulkRNA-seq, Clinical, Imaging | Variants, Genes, Phenotypes | 2021 | N | N | https://www.michaeljfox.org/data-sets | (Padmanabhan et al. 2019) |
Nalls et al. (2019) | Parkinson's | Full genome-wide summary statistics from Nalls et al. (2019) PD GWAS, excluding 23andMe. | GWAS | Variants | 2019 | Y | N | https://pdgenetics.org/resources | (Nalls et al. 2019) |
PD Gene | Parkinson's | Search tool for Nalls et al. (2014) GWAS summary statistics. | GWAS | Variants, Genes | 2017 | Y | N | http://pdgene.org | (Nalls et al. 2014) |
PPMI | Parkinson's | A compilation of PD genomics datasets and tools funded by MJFF via the Parkinson's Progression Markers Initiative (PPMI) initiative. | GWAS, WGS, bulkRNA-seq, Clinical, Imaging | Variants, Genes, Phenotypes | 2019 | Y | N | http://www.ppmi-info.org | (Aleksovski et al. 2018) |
PDBP | Parkinsonianisms | Repository for biosamples and biomarkers data funded by National Institute of Neurological Disorders and Stroke (NINDS) via the Parkinson's Disease Biomarkers Program (PDBP). | GWAS, bulkRNA-seq, Clinical | Variant, Genes, Cell-types, Tissues, Phenotypes | 2021 | N | Y | https://pdbp-demo.cit.nih.gov | (Rosenthal et al. 2016) |
echolocatoR Fine-mapping Portal | Multiple | Multi-tool fine-mapping results and locus visualizations across 11+ GWAS/QTL. | GWAS, QTL | Variants, Genes, Cell-types | 2021 | Y | Y | https://rajlab.shinyapps.io/Fine_Mapping_Shiny | (Schilder et al. ) |
EpiGraphDB | Multiple | Multi-modal epidemiological networks of linking data across phenotypes, literature terms, genes, tissues, drugs and more. ukbiobank-tools provides API access to certain aspects of the database (https://ukbiobank-tools.readthedocs.io/en/latest/index.html). |
GWAS, Literature | Variants, Genes, Cell-types, Tissues, Phenotypes | 2021 | Y | Y | https://www.epigraphdb.org | (Liu et al. 2020) |
EpiMap | Multiple | Expanded and standardised collection of all epigenomics data from ENCODE, the Roadmap Epigenomics Project, and the Genomics of Gene Regulation (GGR). Includes multi-modal networks integrating epigenomics and GWAS. | Epigenomics | Variants, Genes, Cell-types Tissues, Phenotypes | 2021 | Y | Y | http://compbio.mit.edu/epimap | (Boix et al. 2021) |
eQTL Catalogue | Multiple | Uniformly reprocessed, tabix-indexed summary statistics and statistical fine-mapping results from 112 QTL datasets across 21 studies, including QTL for: gene expression, exon expression, transcript usage, and txrevise event usage. | QTL | Variants, Genes, Cell-types, Tissues | 2021 | Y | Y | https://www.ebi.ac.uk/eqtl | (Kerimov et al. 2020) |
FUMA GWAS | Multiple | Web tool for automated GWAS annotation and cell-type/tissue enrichment. | GWAS | Variants, Genes, Cell-types, Tissues, Phenotypes | 2021 | Y | N | https://fuma.ctglab.nl | (Watanabe et al. 2017) |
GenoML | Multiple | Toolkit to facilitate the creation of novel deep learning architectures for genomics data. | GWAS, QTL, Epigenomics, scRNA-seq | Variants, Genes, Cell-types, Phenotypes | 2021 | Y | Y | https://genoml.com | (Makarious et al., 2021) |
Janggu | Multiple | Toolkit to facilitate the creation of novel deep learning architectures for genomics data. | GWAS, QTL, Epigenomics, scRNA-seq | Variants, Genes | 2021 | Y | Y | https://github.com/BIMSBbioinfo/janggu | (Kopp et al., 2020) |
Kipoi | Multiple | Pre-trained deep learning models for genomics data. | GWAS, QTL, Epigenomics, scRNA-seq | Variants, Genes | 2021 | Y | Y | https://kipoi.org | (Avsec et al., 2019) |
MDSGene | Multiple | Databases of variants, genes and symptoms associated with movement disorders in the literature. | Literature | Variants, Genes, Phenotypes | 2020 | Y | N | http://www.mdsgene.org | (Klein et al. 2018) |
MungeSumstats | Multiple | Tool for automatic, robust, and scalable standardization of GWAS summary statistics. Provides API-access to MRC IEU Open GWAS | GWAS | Variants, Phenotypes | 2021 | Y | Y | https://github.com/neurogenomics/MungeSumstats | (Murphy and Skene, 2021) |
OMIM | Multiple | Manually curated database of genes associated with various disease phenotypes. | Literature | Variants, Genes, Phenotypes | 2021 | Y | Y | https://omim.org/entry/168600 | (Hamosh et al. 2005) |
Open GWAS | Multiple | The MRC IEU Open GWAS Project is an online database of semi-standardised, full genome-wide summary statistics from 39k+ GWAS. All GWAS are provided in tabix-indexed VCF format for rapid querying. | GWAS | Variants, Phenotypes | 2021 | Y | Y | https://gwas.mrcieu.ac.uk | (Elsworth et al. 2020) |
Open Targets | Multiple | Query engine to gather variant- and gene-level annotations, GWAS summary statistics, colocalization results, and fine-mapping results. | GWAS, QTL | Variants, Genes | 2021 | Y | Y | https://www.opentargets.org/genetics-portal | (Carvalho-Silva et al. 2019) |
Simple ClinVar | Multiple | Variant annotations including clinical relevance, deleteriousness, and phenotype associations. | GWAS | Variants, Genes, Phenotypes | 2021 | Y | N | http://simple-clinvar.broadinstitute.org | (Pérez-Palma et al. 2019) |
UK Biobank | Multiple | Repository genotyping, imaging, and whole-genome sequencing of deeply phenotyped individuals. | GWAS | Variants, Genes, Phenotypes | 2021 | Y | Y | http://www.nealelab.is/uk-biobank | (Bycroft et al. 2018) |
UK Biobank Correlation | Multiple | Pairwise genetic correlations across all UK Biobank phenotypes. | Phenotype correlations | Phenotypes | 2019 | Y | N | https://ukbb-rg.hail.is | (Bycroft et al., 2018) |
Multi-scale omics: from genome to phenome
Genome-wide association studies (GWAS) have been an invaluable tool to investigate the molecular etiology underlying many complex phenotypes, including PD (Blauwendraat et al., 2020; Chang et al., 2017; Nalls et al., 2019a, 2014a; Parkinson’s Disease Genomics, 2017). To date, there have been over 24 GWAS conducted in PD (Sara Bandres-Ciga et al., 2020; Redenšek et al., 2018) since 2009 (Simon-Sanchez et al., 2009), identifying 78 PD risk-associated loci (Nalls et al., 2019a). Of the known PD risk-associated variants identified through GWAS, the vast majority fall within presumed non-protein-coding regions (Nalls et al., 2019a), posing both additional challenges and opportunities when identifying the genes, pathways, and cell-types via which they exert their effects. Disentangling these systems is complicated by non-independence at multiple biological scales (Table S3A). In addition, one must ultimately consider the developmental, life and/or disease stage at which an underlying genetic risk factor exerts its effects relative to when the patient is being treated (Faa et al., 2014; Kovacs et al., 2014). Underlying all of these goals is the need to first distinguish causal from non-causal risk-associated variants. Thus, the mechanisms underlying known genetic factors are still far from fully understood (Ohnmacht et al., 2020).
Prioritizing putative causal variants
Fine-mapping candidate causal variants within a given GWAS locus is essential as it cannot be assumed that the lead SNP (the one with the smallest p-value) is causal for the phenotype (e.g. PD) due to widespread linkage disequilibrium (LD) (Broekema et al., 2020; Pasaniuc and Price, 2017; Pritchard and Przeworski, 2001). Fine-mapping is a class of statistical tools that aims to identify the causal variants underlying a given phenotype (Benner et al., 2017; Hutchinson et al., 2020; Schaid et al., 2018). Depending on the method, fine-mapping requires either raw genomic sequences or GWAS summary statistics, and may utilize genome-wide functional annotations (e.g. to compute SNP-wise prior probabilities) (Farh et al., 2015; Kichaev et al., 2017; Weissbrod et al., 2020). Parsing out the causal PD risk-associated SNPs from non-causal correlates via fine-mapping is an essential step that can dramatically alter the validity of many downstream analyses which rely upon the identification of the causal sequence motifs. Statistical fine-mapping (without functional annotation input) using FINEMAP (Benner et al., 2016) was recently performed on all loci from the latest PD GWAS summary statistics (Grenn et al., 2020). Additional efforts have been made to statistically fine-map GWAS across many different phenotypes, including PD, via the Open Targets Data Portal (Carvalho-Silva et al., 2019). In a study by our group (Schilder and Raj, 2020), we used a multi-tool consensus fine-mapping approach (Schilder et al., 2020), which included functional fine-mapping of 74/78 known PD GWAS loci using PolyFun (Weissbrod et al., 2020) and cell-type-specific epigenomic variant-annotation, to reduce the average number of putative causal SNPs from 85 to 3 per locus. While the lead SNPs were indeed within the 95% probability union credible set (nominated by at least one tool) for 69/74 (93.2%) loci, they were in the consensus SNP set (nominated by at least two tools) in only 29/74 (39.2%) of loci. This emphasizes the need for robust fine-mapping to disambiguate causal variants from their close correlates. Two specific examples of consensus fine-mapped SNPs from this study include rs7294619 (LRRK2 locus), which disrupts LRRK2 expression via a FCGR2A TFBM in a microglia-specific enhancer, and rs4771268 (MBNL2 locus), which disrupts an MBNL2-regulating enhancer in oligodendrocytes alone. Fine-mapping results and figures from all of these studies are freely available via their respective online data portals (Table 1).
In recent years, machine learning (ML) has been especially successful at computationally modeling the functional genome, thus improving the accuracy and precision of the functional impact of mutations (Avsec et al., 2021, 2019; Eraslan et al., 2019; Kelley et al., 2018; Zhou and Troyanskaya, 2015). Dey et al. (2020) have provided genome-wide predictions from multiple deep learning models (e.g. DeepSEA (Zhou and Troyanskaya, 2015), Basenji (Kelley et al., 2018), ExPecto (Zhou et al., 2018), and Enformer (Avsec et al., 2021)) for all possible genomic mutations through a process called in silico mutagenesis. These predictions are of particular relevance to PD (Schilder and Raj, 2020) as they have trained separate models on blood- or brain-specific data. In an effort to facilitate research and minimize redundant efforts, we have compiled a table of existing resources relevant to PD (Table 1). We’d like to note several particularly relevant and useful resources for making powerful ML tools accessible to non-experts, including GenoML (Makarious et al., 2021), Kipoi (Avsec et al., 2019), and Janggu (Kopp et al., 2020).
Prioritizing putative causal genes
There are several main approaches used when inferring the genes that non-coding variants may regulate: proximity (usually based on transcription start site), quantitative trait loci (QTL) (Falconer, 1996; Kearsey, 1998), Transcriptome-Wide Association Studies (TWAS) (Gamazon et al., 2015; Gusev et al., 2016), and 3D chromatin interactions (Bhattacharyya et al., 2019; Fulco et al., 2019; Juric et al., 2019; Nasser et al., 2021; Sey et al., 2020). Many investigations of PD have used the proximity-based strategy Multi-marker Analysis of GenoMic Annotation (MAGMA) (de Leeuw et al., 2015). However, disease-associated SNPs frequently regulate the gene (or genes) that are not the one closest to the lead (or even causal) SNP (Fulco et al., 2019; Garrido-Martín et al., 2021; Mumbach et al., 2017; Nasser et al., 2021). Thus proximity alone is not necessarily a reliable proxy for variant-gene interactions. Identifying causal genes in GWAS loci is especially challenging as these regions tend to be gene-dense. In the case of PD, there are 305 genes within the 78 known risk loci (±1Mb from the lead SNPs) (Nalls et al., 2019a),
One approach to prioritize putative causal genes is to incorporate information from expression or splicing QTLs. Early attempts used looked for overlap between PD-associated SNPs from several GWAS (Lill et al., 2012) and QTLs in GTEx (Coetzee et al., 2016; Pierce and Coetzee, 2017a). However, as described above, LD confounds the accurate identification of causal SNPs in both GWAS and QTL studies. To identify colocalization of GWAS and eQTL associations, Raj et al. (2014a) applied Regulatory Trait Concordance (RTC) scores (Nica et al., 2010) in monocytes datasets to identify shared eQTL and PD GWAS signals in seven PD GWAS loci (including LRRK2, BST1, and SNCA). Others have used Bayesian colocalization tests (e.g. with the tool coloc) (Giambartolomei et al., 2014) to assess whether the genetic signals underlying GWAS and QTL in a particular genomic region are shared, thereby mitigating the confounding effects of LD. This has been applied to PD in multiple studies using partially overlapping QTL reference datasets (Li et al., 2019; Schilder and Raj, 2020). This is a much more robust approach than simply checking whether a specific genome-wide significant GWAS SNP is also a significant QTL hit (confusingly, also often referred to as “colocalization” when the term “overlap” would be more appropriate). More recently, as part of the Myeloid Cell Neurodegenerative disease (MyND) project, Navarro and Udine et al. (2020) observed that in 17 PD susceptibility loci, the PD risk variants are likely to modify disease susceptibility, at least in part, by modulating gene expression or splicing in peripheral monocytes. In another study, using Microglia Genomic Atlas (MiGA), (de Paiva Lopes et al., 2020)Lopes et al. (2020) demonstrated 18 PD GWAS loci colocalize with microglia eQTLs including P2RY12, which is highly expressed in microglia in comparison to other brain and myeloid cell types and has been shown to play a role in microglia migration and activation.
Mendelian Randomization (MR) is a statistical methodology used to infer the causal relationships between multiple genotype-phenotype association datasets (Yang et al., 2021). Nalls et al. (2019b) applied MR to the GWAS, expression QTLs and methylation QTLs from multiple cell types and tissues to narrow the potential candidates to 150 genes. They found that one of the putative causal genes was the one nearest to the putative causal SNP in 53/70 (76%) of PD GWAS loci. However, most loci contained multiple putative causal genes according to this approach. More extensive gene nomination was later performed using MR on fine-mapped GWAS loci and blood/brain eQTLs, in combination with a literature-based gene-scoring strategy (Grenn et al., 2020).
Another approach to prioritize causal genes in GWAS loci is to directly test for association between a disease and gene expression. However, such study is currently not feasible since it requires profiling gene expression across hundreds of thousands of individuals in cases and controls. An alternative approach is to perform TWAS, which leverage information from GWAS and eQTL datasets to impute (or predict) the gene expression of cases and controls, thus allowing the direct association of disease and genes without directly profiling gene expression in every individual ((Gamazon et al., 2015; Gusev et al., 2016). Several TWAS of a number of neurological disorders have been performed, incorporating a variety of source tissues and cell types (Gusev et al., 2018; Raj et al., 2018). With respect to PD, Li et al. (Li et al., 2019) performed TWAS using bulk transcriptomes across multiple tissues from GTEx (GTEx Consortium, 2015; Pers et al., 2015), as well as prefrontal cortex (Fromer et al., 2016), and immune cells (Fairfax et al., 2014; Heng et al., 2008; Raj et al., 2014a; Rotival et al., 2011) to nominate 66 PD-related genes, as well as specific transcripts, which can have more tissue- and cell-type-specific effects via alternative splicing. We do note that there are limitations to these gene prioritization methods. TWAS (as well as colocalization and MR-based methods) are prone to gene co-regulation bias, where the causal gene has correlated total expression with another neighboring gene, which can lead to a false-positive TWAS association (see Wainberg et al. (2019) for further discussion).
Lastly, 3D genomic information, usually in the form of chromatin interaction maps, can be utilized to infer SNP-gene relationships, using Chromosome Conformation Capture coupled with sequencing (HiC) maps (Lieberman-Aiden et al., 2009) (e.g. H-MAGMA) (Sey et al., 2020), HiChIP (Mumbach et al., 2016) and/or Proximity Ligation-Assisted ChIP-seq (PLAC-seq) (Fang et al., 2016; Park, 2009) (e.g. Model-based Analysis of PLAC-seq (MAPS) (Juric et al., 2019; Nott et al., 2019; Schilder and Raj, 2020) and FitHiChIP (Bhattacharyya et al., 2019; Corces et al., 2020)), and Activity-by-Contact (ABC) maps from CRISPRi-FlowFISH (Fulco et al., 2019; Nasser et al., 2021). The availability of large-scale, harmonised databases like EpiMap (Boix et al., 2021) will permit researchers to fully utilize these tools. This approach is promising due to its strong basis in mechanistic understanding of genome regulation and direct experimental measurement, but is currently limited by the fact that chromatin interaction maps differ across different cell-types, which are difficult to disentangle from bulk data alone.
Nevertheless, even these more sophisticated methods can be subject to false positive associations and are highly dependent on the availability and selection of QTL/epigenomic datasets in relevant tissues and cell-types. Varying sample sizes can also falsely make well-powered datasets (e.g. eQTLs in blood) appear to be causal (Grenn et al., 2020; Pierce and Coetzee, 2017b). Furthermore, most studies have only considered cis-QTLs due to the multiple testing burden intrinsic in trans-QTL studies, though some recent advances have been made to reduce this burden through tensor decomposition (Ramdhani et al., 2020). Therefore, care and expertise are required when attempting to integrate GWAS and QTL data for SNP or gene prioritization. The development of new technologies and the generation of new datasets that resolve chromatin interaction maps at single cell resolution will be invaluable moving forward.
Identifying pathogenic cell-types and tissues
In addition to DA neurons, a wide variety of cell-types has been implicated in the genetic etiology of PD. Nalls et al. (2019c) tested for enrichment of PD GWAS-inferred genes across 88 brain cell-types in a mouse single-cell RNA-seq (scRNA-seq) reference. The strongest enrichment was in neurons of the SN, globus pallidus, thalamus, posterior cortex, frontal cortex, hippocampus, and entopeduncular nucleus. Similarly, others have also found enrichment for various neuronal subtypes including nigrostriatal DA neurons (using a human transcriptomic reference) (Agarwal et al., 2020) and their parent class monoaminergic neurons (using a mouse transcriptomic reference) (Bryois et al., 2020a), cortical excitatory neurons, as well as cortical inhibitory neurons (Agarwal et al., 2020; Li et al., 2021). Other studies have implicated various glial cell-types, including oligodendrocytes (Agarwal et al., 2020; Bryois et al., 2020b; Nott et al., 2019), oligodendrocyte precursor cells (OPCs) of the SN (but not the cortex) (Agarwal et al., 2020), astrocytes (Kam et al., 2020), and microglia (Andersen et al., 2021; Badanjak et al., 2021; de Paiva Lopes et al., 2020; Ho, 2019; Kam et al., 2020). Genes that are differentially expressed across Braak stages in post-mortem PD brain samples (Dijkstra et al., 2015) were also enriched for transcriptional signatures for DA neurons and oligodendrocytes (Bryois et al., 2020a). Interestingly, some evidence suggests that LRRK2 is more highly expressed in nigral OPCs than any other cell-type in that region (Agarwal et al., 2020), though replication with more individuals is needed. One hypothesis regarding the role of microglia is that they serve to clear α-synuclein through phagocytosis (Ho, 2019; Poewe et al., 2017), though other studies could find no significant enrichment for microglia in PD GWAS (Agarwal et al., 2020; 2020a; Reynolds et al., 2019). Additional candidate cell-types within the peripheral immune system have also been proposed, including lymphocytes (Coetzee et al., 2016), T-cells (Garretti et al., 2019), monocytes (Navarro et al., 2020; Raj et al., 2014a) and macrophages (de Witte et al., 2020). Some studies have even implicated enteric neurons (Bryois et al., 2020b), mesoderm, liver and fat cells (Coetzee et al., 2016). Lastly, some have found a lack of genome-wide PD heritability enrichment for any particular cell-type, suggesting the mechanism was at the level of cell-type-agnostic processes such as lysosomal function (Reynolds et al., 2019). This is despite the fact that they did indeed find cell-type-specific enrichment for schizophrenia heritability, and used a highly similar methodology to another study that found enrichment for several tissues and cell-types in PD heritability (Bryois et al., 2020a). Similarly, others have found no genome-wide enrichment of cell-type-specific epigenomic signatures in PD GWAS after multiple testing corrections across multiple phenotypes (Nott et al., 2019).
The source of these seemingly inconsistent genetic enrichment results could be both biological and methodological in origin. In particular, differing genetic enrichment tool types, versions and parameters can capture different aspects of the PD etiology and thus have dramatic impacts on conclusions, e.g.; genome-wide LD SCore regression (LDSC) (Finucane et al., 2015), or gene-level enrichment using MAGMA and Expression Weighted Cell-type Enrichment (EWCE) (Bryois et al., 2020a; Skene and Grant, 2016). We elaborate on these non-mutually exclusive possibilities in Table S3C-D. Another important distinction to make is between studies that use cell-type references derived from a small number of individuals or animals (e.g. (Agarwal et al., 2020; 2020a; Corces et al., 2020; Nott et al., 2019; Reynolds et al., 2019; Schilder and Raj, 2020)) and those that use population-scale cell-type references (de Paiva Lopes et al., 2020; Navarro et al., 2020; Young et al., 2021).
Rather than testing for enrichment of particular cell-types genome-wide, a complementary approach is to systematically investigate locus-specific mechanisms. Similar to some prior studies (Nott et al., 2019; Reynolds et al., 2019), Corces et al. (2020) found no genome-wide enrichment for any major brain cell-type or neuronal cell-subtype based on chromatin accessibility peaks in PD, despite finding enrichment for other brain-related phenotypes (e.g. schizophrenia). In contrast, when they systematically interrogated each PD GWAS locus using cell-type-specific epigenomics (scATAC-seq) and interactomics (HiChIP and PLAC-seq), they were able to identify putatively causal mechanisms. For example, within the ITIH1 PD locus they identify a fine-mapped SNP (rs18139313) whose PD-associated allele falls within a KLF4 transcription factor binding motif (TFBM), and interacts with the transcription start site (TSS) of the gene STAB1 exclusively in microglia. Prior to this, there was no known mechanism by which this locus influenced PD risk. Using a similar per-locus approach, Schilder & Raj (2020) found that fine-mapped consensus SNPs fell within a single cell-type (neurons, astrocytes, oligodendrocytes or microglia) in only 14/74 loci (19%), two cell-types in 12 loci (16%), three cell-types in 10 loci (14%), and all four cell-types in 9 loci (12%). Fine-mapped SNPs most frequently fell within microglia peaks, followed by neurons (including nigral neurons) and then oligodendrocytes/oligodendrocyte precursor cells. Of course, the per-locus approach comes with its own set of drawbacks (e.g. availability of multi-omics data in the relevant cell-types) and should be considered complementary to genome-wide enrichment strategies.
Very recently, single-cell and single-nuclei datasets from PD patient samples have begun to emerge, as opposed to the majority of existing datasets which come from mice or humans without PD. Early results indicate that microglia, astrocytes and oligodendrocytes are differentially activated relative to patients with multiple systems atrophy (MSA) and/or controls (Pande et al., 2021; Teeple et al., 2021). Remarkably, these genes were enriched for PD GWAS genes. MSA is a useful comparator as it allows researchers to determine whether the observed effects are specific to PD or more broadly indicative of neurodegenerative disease. Single-nucleus datasets from non-PD post-mortem brain samples are also valuable resources to indirectly explore PD-related questions, including some done in SN (n = 5 individuals) (Agarwal et al., 2020), midbrain DA neurons (Kamath et al., 2021), frontal cortex (Langston et al., 2021b), developing human brain (Bocchi et al., 2021; Cao et al., 2020; Domcke et al., 2020). Bulk cell-type purified RNA-seq in peripheral monocytes (n = 230 individuals) (Navarro et al., 2020) and microglia (n = 55 PD individuals) (de Paiva Lopes et al., 2020) from PD patients has revealed increased expression of mitochondrial related pathways relative to age-matched controls. Currently there is a lack of population-scale datasets in SN or striatum that are sufficiently well-powered to reliably perform analyses such as QTL, much less in cohorts of PD patients. Existing post-mortem studies from SN all have 16 or fewer PD cases (Lesnick et al., 2007; Moran et al., 2006). These datasets allow for comparisons of cell-type-specific signatures, opening up new possibilities for more deeply understanding the cell-type-specific mechanisms underlying PD. Efforts to gather population-scale single-cell QTL datasets are currently underway (van der Wijst et al., 2020).
Considering the phenome
PD in the absence of any other illness is exceedingly rare given the age demographic of patients and the ubiquity of pleiotropy (Li et al., 2017). Therefore, there is a strong impetus to conceptualize human health as a continuous multi-dimensional spectrum. Indeed, within-patient clinical presentations of PD can evolve over time depending on life stage, comorbidities, medications and environmental exposures. Thus, it can be useful to think of PD not as a singular diagnosis, but as a constellation of sub traits (e.g. symptoms) that can each be linked to specific genetic and cellular mechanisms. Methods designed to explore phenome-wide interactions include epidemiological comorbidity observations, genetic correlation using heritability estimates, fine-mapped causal variant overlap, MR, dimensionality reduction (Tanigawa et al., 2019), and multi-trait Polygenic Risk Scores (PRS) (Aguirre et al., 2021). Indeed, a study of 17 autoimmune diseases GWAS revealed extensive genetic sharing (i.e. pleiotropy) between these conditions and PD, providing a causal mechanism underlying the longstanding observation of comorbidity between these conditions (Witoelar et al., 2017). The possibility of large-scale multi-phenotype studies has become increasingly feasible with the advent of databases such as the OpenGWAS (Elsworth et al., 2020) and the Global Biobank Engine (McInnes et al., 2019), as well GWAS summary statistics standardisation pipelines like MungeSumstats (Murphy and Skene, 2021). In Table S2, we provide correlations between PD and many other phenotypes compiled across multiple studies and databases (Bryois et al. 2020; Nalls et al. 2019; Agarwal et al. 2020), allowing for the re-analysis of previously reported relationships and the discovery of novel ones.
Biological pathways
GWAS and multi-omics approaches have been invaluable for the discovery of causal variants, genes, and cell types underlying PD. However, merging these findings into a cohesive, mechanistic understanding of the pathways they are part of is especially challenging. Protein-Protein-Interaction (PPI) (Pintacuda et al., 2021) and multi-modal (Sara Bandres-Ciga et al., 2020; Boix et al., 2021; Greene et al., 2015; Liu et al., 2020; Wang et al., 2019; Wong et al., 2018) networks have often been used to link these components. However, these approaches often suffer from an over-saturation of interactions that obscure relevant signals, as well as limited actionability. Here, we highlight several pathways with some of the strongest evidence for their role in the pathogenesis of PD: endolysosomal/protein degradation, immune, and mitochondrial.
Endolysosomal pathway and protein degradation
Endolysosomal pathways encompass a number of processes essential for cell homeostasis. Lysosomes degrade structurally diverse substances, not only proteins but also nucleic acids, lipids, helped by the acidic environment inside these organelles mainly driven by the V-ATPase proton pump and the presence of a number of proteases which are active at acidic pH. Lysosomes receive cargo from autophagic and endocytic pathways, which deliver substrates from either intracellular or extracellular compartments, respectively. Both processes require the formation, transport and fusion of vesicles, with extensive crosstalk and shared mechanisms (Birgisdottir and Johansen, 2020). Of direct relevance to PD, the intracellular homeostasis of α-synuclein is maintained by the actions of the ubiquitin–proteasome system and the Lysosomal Autophagy System (LAS).
The discovery that LRRK2 has a specific role on vesicular trafficking and GBA in lysosomal function, in addition to the fact that a number of monogenic forms of PD affect autophagy genes, implicate this pathway in the pathogenesis of the disease. Moreover, several independent approaches have demonstrated that this pathway is also enriched for genetic risk in idiopathic forms of PD. Given that GBA mutations can also cause Gaucher disease, which is indeed a lysosomal storage disorder (LSD), Robak et al showed association signals between LSD damaging variants and PD participants (Robak et al., 2017). Applying PRS to 252 endosomal membrane trafficking pathway genes showed their cumulative contribution to PD (Bandres-Ciga et al., 2019). In a broader investigation of pathway enrichment in PD, the vesicle transport pathway was significantly associated with PD risk genetics (S. Bandres-Ciga et al., 2020). Others have used single cell RNA-seq from cortical and SN samples in PD donors to reconstruct cell-cell interactions, identifying pathways such as proteasome, autophagy, endocytosis or vesicle-mediated pathway (Agarwal et al., 2020). Moreover, transcriptomic data generated in human fresh microglia and monocytes from idiopathic cases of PD showed a downregulation of proteo-lysosomal genes (Navarro et al., 2020).
Mitochondria
Dysfunctional mitochondrial activity has a well-established role in the pathology of PD (Poewe et al., 2017). Evidence for this primarily comes from observations in environmentally induced mitochondrial blockades (Williams, 1984), and histopathological investigations of mitochondrial complex I disruption in postmortem brains of patients with idiopathic PD (Mizuno et al., 1989; Schapira et al., 1990). That said, mitochondrial dysfunction is also a major pathway altered in monogenic forms of PD, specifically those associated with PRKN/PINK1 missense mutations (Padmanabhan et al., 2019; Pickrell and Youle, 2015). Yet the putative role of common PD-associated variants in mitochondrial pathways has been more difficult to define. Interestingly, researchers calculated a mitochondrial PRS and demonstrated that common variants within mitochondrial pathway genes are associated with PD disease status (Billingsley et al., 2019). In an independent line of evidence, PD susceptibility variants within mitochondrial pathway genes were identified using Whole-Genome Sequencing (WGS) and subsequently confirmed using functional validation experiments (Jansen et al., 2017). In a transcriptomic characterization of human myeloid cells in PD, we showed how there is a significant dysregulation in genes associated to oxidative phosphorylation, even though the directionality of the effect seems to be flipped in monocytes vs microglia/macrophages (Navarro et al., 2020). Other authors have also shown mitochondrial alterations in human-derived cells (Annesley et al., 2016; Mortiboys et al., 2010; Teves et al., 2017). Thus, mitochondrial pathways play a role in PD etiology at multiple layers.
Finally, it is worth mentioning that the mitochondrial pathway is not independent of other pathways implicated in PD. For example, the autophago-lysosomal pathway is necessary to remove damaged mitochondria, and its dysfunction increases the accumulation of aberrant organelles which contributes to pathology. This can lead to deleterious feedback loops between lysosomes and mitochondria (Park et al., 2018).
Immune system
Beyond the neuronal component, immune cells have also been associated with the development of PD through the identification of several common genetic variants within genes related to immune response (e.g. LRRK2, TNF, HLA-DRB5) (Chu et al., 2012; Hamza et al., 2010; Paisán-Ruíz et al., 2004; Zimprich et al., 2004). Additional evidence suggests PD heritability enrichments for epigenomic marks (including DNase I hypersensitive sites, histone modifications, and transcription factor binding sites) in both adaptive and innate immune cells (Gagliano et al., 2016) and a number of PD-associated genes are preferentially expressed in microglia (Gosselin et al., 2017). Raj et al. (2014b) demonstrated that monocyte eQTLs are enriched for PD GWAS signal, suggesting a functional role of the innate immune system into the development of PD. Additionally, a TWAS of PD found 29 loci mediated through either mRNA expression or splicing in monocytes to be associated with PD, including both LRRK2 and SNCA (Li et al., 2019). Studies performed in human-derived monocytes have shown increased reactivity, metabolic alterations and increased levels of LRRK2 (Cook et al., 2017; Grozdanov et al., 2019, 2014; Schlachetzki et al., 2018; Smith et al., 2018). A population-based dataset of freshly isolated monocytes from idiopathic cases of PD showed profound dysfunction in the peripheral immune system, further solidifying the pivotal role of these cell types (Navarro et al., 2020).
In addition to myeloid cells, recent findings have also implicated the adaptive immune system in causal PD pathology. In particular, there is evidence of T-cell infiltration in postmortem PD brains (Brochard et al., 2009) and α-synuclein driving cytotoxic responses in T-cells. New datasets of T-cells derived from PD patients are needed to further elucidate the role of these cells in the disease.
The inflammatory component in PD has been corroborated through multiple observations in humans, including the presence of reactive microglia in post-mortem brains (Banati et al., 1998; Imamura et al., 2003; McGeer et al., 1988; Mirza et al., 2000), increased cytokine levels in plasma (Dobbs et al., 1999; Rentzos et al., 2007; Stypuła et al., 1996) and cerebrospinal fluid (CSF) (Mogi et al., 1995, 1994), as well as microgliosis investigated using Positron Emission Tomography (PET) imaging (Gerhard et al., 2006). All these findings support the hypothesis that both, microglia (resident immune cells in the central nervous system) as well as peripheral monocytes and macrophages are playing a role in the development of this disease (Kannarkat et al., 2013; Tansey and Romero-Ramos, 2018). Together, these studies are beginning to provide a mechanistic understanding of the frequently observed comorbidity between PD and immune disorders (Arai et al., 2006; Li et al., 2012; McGeer and McGeer, 2004; Phani et al., 2012; Witoelar et al., 2017).
Experimental validation
Although analysis of human samples is useful for hypothesis generation, validating causal relationships between proposed mechanisms and disease risk/progression requires a degree of experimental control that would be logistically infeasible (e.g. sample collection and processing) and/or unethical in human patients (e.g. untested drug screens or genetic perturbations). As PD susceptibility appears to be a human-specific trait (Garcia-Ruiz and Espay, 2017) , animal models that can reliably recapitulate key features of PD are currently lacking (Potashkin et al., 2010). Thus, new experimental tools are needed to validate and further understand the functional consequences of PD-associated variants. Here we review some of the most prominent experimental in vitro methodologies and results that have been used to validate PD GWAS-nominated risk factors. For reviews of animal models in PD research, please see the following: (Jagmag et al., 2015). While there is a much more extensive literature on validation of rare PD risk factors (Poewe et al., 2017), we primarily focus on methods and results pertaining to common PD risk variants and the exciting opportunities that lay ahead in this area.
In vitro modeling
High-throughput technologies are emerging to validate disease-associated variants and understand their functional consequences. Induced pluripotent stem cells (iPSCs) can be generated from patient tissue samples and thereafter differentiated into a variety of cell-types (Takahashi and Yamanaka, 2006), opening up a wide range of possibilities fo PD research (Coccia and Ahfeldt, 2021). One major advantage of patient-derived iPSCs over animal models is that they are able to capture the genome-wide complexity of human disease genetics, as opposed to one or several artificially-introduced genetic perturbations amidst a non-human genomic background. Thus, iPSC-derived DA neurons from PD patients can be compared to those of healthy, matched controls (Soldner et al., 2009). Taking this approach, several studies have shown that patient-derived DA-neurons recapitulate pathogenic events such as loss of dendritic arborization and autophagy defects (Sánchez-Danés et al., 2012), epigenomic dysregulation (Fernández-Santiago et al., 2015), microRNA and piRNA alterations (Schulze et al., 2018; Tolosa et al., 2018) and hypermethylation of regulatory regions (Fernández-Santiago et al., 2019). Another is to stratify iPSC-derived cells by PRS, as a way to move from genetic risk to phenotypic characterization.
However, increased genomic complexity and variation of models also mean that small, context-dependent effects are difficult to detect, especially when sample sizes are limited due to practical or budget constraints. A powerful complementary functional validation approach uses CRISPR-Cas9 (Adli, 2018; Cong et al., 2013) to induce targeted genetic editing of prioritized variants in isogenic human iPSC-derived cells, allowing researchers to directly observe the consequences of specific variants while keeping genetic background constant. Of special help are new versions of CRISPR which use a dead-Cas9 and allow to either activate gene expression (CRISPRa) or repression (CRISPRi), rather than completely knocking out genes. Gene editing has been widely used to model rare monogenic PD mutations (Burbulla et al., 2017; Fong et al., 2013; Ryan et al., 2013; Soldner et al., 2011), but has far more rarely been applied to common PD variants. One of the earliest examples comes from Soldner et al. (2016), in which they targeted a common variant in a non-coding distal enhancer and demonstrated a significant effect on the expression of SNCA, one of the most consistent genetic risk genes in PD. Others have focused on common variants associated with LRRK2 and have modelled either in iPSC derived neurons (Marrone et al., 2018) or microglia (Langston et al., 2021a). We (de Paiva Lopes et al., 2020; Schilder and Raj, 2020) and others (Langston et al., 2021a) have demonstrated that microglia-specific regulatory regions that may modulate LRRK2 expression but Langston et al. (Langston et al., 2021a) found no difference using CRISPR-Cas9 editing from protective to risk variant (for the lead GWAS SNP, rs76904798) in iPSc microglia. It is plausible that the lead SNP is not the causal variant but another variant (rs7294619) may be causal at the LRRK2 locus.
Rather than focusing on one variant at a time, new technologies are being developed to perform combinatorial CRISPR-based genetic perturbation screens in iPSCs, permitting the exploration of both additive and interactive effects of multiple variants at once(Fernando et al., 2020). Even greater resolution can be achieved by combining these methods with single-cell RNA-seq, as in Perturb-seq (Dixit et al., 2016), allowing for the investigation of cell-specific effects of genetic modifications on gene expression. This approach has been applied to a variety of studies such as transcriptional regulation of unfolded protein response (Adamson et al., 2016), neuronal development (Alyagor et al., 2018) or autism risk genes (Jin et al., 2020). In PD research, a knock-out CRISPR-Cas9 screen was developed to assess genetic regulators of the gene PRKN (Potting et al., 2018). Another example is PRISM (Perturbing Regulatory Interactions by Synthetic Modulators), a platform that edits random transcription factors (TFs) with CRISPR-Cas9 to assess transcriptional networks that protect against α-synuclein in yeast (Chen et al., 2017). Moving forward, these approaches are expanding to the use of CRISPRa (CRISPR activation), CRISPRi (CRISPR interference) and gene editing, and will help to disentangle the genetics of polygenic diseases such as PD. In this line, Tian and collaborators recently published genome-wide CRISPRi and CRISPRa screens in human-derived neurons, where they uncover molecular pathways related to lysosomal failure and oxidative stress, implicating the PD-related genes FBXO7 and PSAP (Tian et al., 2021).
A complementary unbiased approach for discovering and/or verifying disease-associated variants is the use of massively parallel reporter assays (MPRAs), which assess the functional impact of thousands of variants and sequences in parallel. This high-throughput technology has traditionally been used to study the “regulatory grammar” and it is extensively reviewed in the context of neuropsychiatric disorders elsewhere (Mulvey et al., 2021; Townsley et al., 2020). This approach permits comparisons of the activity in transfected vs reference cells, allowing the identification of regulatory elements in a cell and context specific manner. It has been applied to assess thousands of human polymorphisms with the identification of up to 842 variants which affect gene expression across different GWAS traits, including the regulation in the expression of STX4 in PD (Tewhey et al., 2018).
Although iPSCs are an invaluable tool for studying disease mechanisms and expediting drug discovery, it has several key limitations at present. High variability (between cell lines and between laboratories), makes comparisons across research groups or studies extremely challenging. Therefore, improved experimental protocols, data harmonization, and in silico data integration will be pivotal moving forward. In addition, these experiments are expensive and labor intensive. Nor do they currently account for the epigenetic changes upon culture, the lack of maturation in some cases, or the fact that these cells are isolated in a dish as opposed to integrated into a living system. That said, organoids generated from stem cells may be better able to recapitulate certain aspects of human biology, such as the integration of multiple cell types in the same model, and becoming self-organized in a 3D structure. Of especial relevance, midbrain organoids may be able to better model the orchestrated behavior of multiple cell types in PD pathogenesis (Galet et al., 2020; Kim et al., 2019; Smits et al., 2019). In addition, part of the iPSC Neurodegeneration Initiative (INDI) is planning to generate publicly accessible isogenic iPSC carrying PD-relevant mutations (Riley and Schekman, 2021), which will serve as an important resource for researchers which will also help to harmonize iPSC PD models.
Future directions and conclusions
Various large-scale initiatives are currently underway to generate or add to multi-omics datasets for research into PD more generally (Table 1). Some notable examples include the Parkinson's Progression Markers Initiative (PDBP) (Rosenthal et al., 2016), the Accelerating Medicine Partnership: Parkinson’s Disease (AMP PD) (Iwaki et al., 2020), the Foundational Data Initiative for Parkinson's Disease (FOUNDIN-PD) (Bressan et al., 2021), Aligning Science Across Parkinson (ASAP) (Schekman and Riley, 2019), and the Global Parkinson’s Genetics Program (GP2) (The Global Parkinson’s Genetics Program, 2021). The UK Biobank’s (UKB) is a more general resource containing whole-genome sequencing data from 500,000 deeply phenotype individuals (Bycroft et al., 2018). Similarly, more work is needed to conclusively resolve the causal cell-types underlying PD, though progress is being made by consortia such as the Human Cell Atlas (HCA) (Regev et al., 2017) and single-cell eQTLGen (van der Wijst et al., 2020). Furthermore, it should be noted that while identifying the cell-types that causally underlie a disease is undoubtedly an informative and worthwhile pursuit, these may not necessarily be the ideal candidate cell-types to directly target with therapeutics. Additional factors that first need to be considered are the delivery method and the potential for off-target effects. To fully utilize such resources, we also need well-designed, programmatically-accessible, cloud-based architectures, improved data sharing standards and novel computational methods.
Lastly, we would like to emphasize that the approaches laid out here should be in no way construed as a comprehensive list of all the ways in which PD may be investigated. The various subfields of biomedicine are constantly evolving at a rapid pace, whilst producing data at a rate that lends itself to underutilization. Therefore, novel innovative experimental designs, computational tools, and conceptual frameworks in the years to come will be essential to tackle the formidable challenges that arise when dissecting the complex diseases like PD.
Supplementary Material
Acknowledgements
We would like to thank Jack Humphrey, Gijsje Snijders, Elena Coccia, Giulietta Maria Riboldi, Katia de Paiva Lopes, and Ricardo Vialle for their invaluable feedback on this manuscript. We would also like to thank the following people for their contributions of cell-type/tissue enrichment summary statistics: Mike Nalls, Cornelis Blauwendraat, Nathan Skene, Caleb Webber, Cynthia Sandor and Alexi Nott.
This work is dedicated to Robert Neil Cronin.
Funding
This work was supported by grants awarded to TR by the Michael J. Fox Foundation (Grant #14899 and #16743) and US National Institutes of Health (NIH NIA R01-AG054005, NINDS R01-NS116006 and NINDS U01-NS120256). The author BMS was additionally supported by the UK Dementia Research Institute which receives its funding from UK DRI Ltd, funded by the UK Medical Research Council, Alzheimer's Society and Alzheimer's Research UK.
Glossary
Acronym | Definition | Type | Reference |
---|---|---|---|
SN | Substantia Nigra | anatomy | |
ChIP-seq | Chromatin ImmunoPrecipitation and sequencing | assay | (Park 2009) |
CRISPRa | CRISPR activation | assay | |
CRISPRi | CRISPR interference | assay | |
CRISPRi-FlowFISH | CRISPRi, RNA fluorescence in situ hybridization (FISH), and flow cytometry | assay | (Nasser et al., 2021) |
HiC | Chromosome Conformation Capture (3C) coupled with sequencing | assay | (Mumbach et al. 2016) |
HiChIP | portmanteau of HiC and ChIP-seq | assay | (Lieberman-Aiden et al. 2009) |
Perturb-seq | portmanteau of "perturbations" and "sequencing" | assay | (Dixit et al., 2016) |
PLAC-seq | Proximity Ligation-Assisted ChIP-seq | assay | (Fang et al. 2016) |
PRISM | Perturbing Regulatory Interactions by Synthetic Modulators | assay | (Chen et al., 2017) |
RNA-seq | RiboNucleic Acid sequencing | assay | |
scATAC-seq | single-cell Assay for Transposase-Accessible Chromatin with sequencing | assay | |
scRNA-seq | single-cell RiboNucleic Acid sequencing | assay | |
WGS | Whole-Genome Sequencing | assay | |
DA neurons | DopAminergic neurons | cell-type | |
OPCs | Oligodendrocyte Precursor Cells | cell-type | |
AMP PD | Accelerating Medicine Partnership: Parkinson’s Disease | consortium | (Iwaki et al., 2020) |
ASAP | Aligning Science Across Parkinson | consortium | (Schekman and Riley, 2019) |
FOUNDIN-PD | Foundational Data Initiative for Parkinson's Disease | consortium | (Bressan et al., 2021) |
GTEx | Genotype-Tissue Expression | consortium | (GTEx Consortium, 2015; Pers et al., 2015) |
HCA | Human Cell Atlas | consortium | (Regev et al., 2017) |
INDI | iPSC Neurodegeneration Initiative | consortium | (Riley and Schekman, 2021) |
UKB | UK Biobank | consortium | (Bycroft et al., 2018). |
MiGA | Microglia Genomic Atlas | dataset | (Lopes et al., 2020) |
MyND | Myeloid Cell Neurodegenerative disease | dataset | (Navarro et al., 2020) |
LSD | Lysosomal Storage Disorder | disease | (Robak et al., 2017) |
MSA | Multiple Systems Atrophy | disease | (Pande et al., 2021; Teeple et al., 2021) |
PD | Parkinson's Disease | disease | (Parkinson 1817) |
Cas9 | CRISPR-associated protein 9 | feature | |
DNase | DeoxyriboNuclease | feature | |
LAS | Lysosomal Autophagy System | feature | (Birgisdottir and Johansen, 2020) |
LD | Linkage Disequilibrium | feature | |
piRNA | Piwi-interacting RNA | feature | |
PPI | Protein-Protein-Interaction | feature | (Pintacuda et al., 2021) |
SNP | Single-Nucleotide Polymorphism | feature | |
TF | Transcription Factor | feature | |
TFBM | Transcription Factor Binding Motif | feature | |
TSS | Transcription Start Site | feature | |
V-ATPase | Vacuolar-Type Adenosine Triphosphate -ase | feature |
ABC | Activity-by-Contact | methodology | (Fulco et al., 2019; Nasser et al., 2021) |
CRISPR | Clustered Regularly Interspaced Short Palindromic Repeats | methodology | |
eQTL | expression Quantitative Trait Loci | methodology | |
GWAS | Genome-Wide Association Study | methodology | |
iPSCs | induced Pluripotent Stem Cells | methodology | |
machine learning | ML | methodology | |
MPRA | Massively Parallel Reporter Assay | methodology | (Mulvey et al., 2021; Townsley et al., 2020) |
MR | Mendelian Randomisation | methodology | |
PRS | Polygenic Risk Score | methodology | |
QTL | Quantitative Trait Loci | methodology | |
RTC | Regulatory Trait Concordance | methodology | |
tQTL | transcript Quantitative Trait Loci | methodology | |
TWAS | Transcriptome-Wide Association Study | methodology | |
Basenji | Basenji | tool | (Kelley et al. 2018), |
DeepSEA | Deep learning–based SEquence Analyzer | tool | (Zhou and Troyanskaya 2015) |
Enformer | portmanteau of enhancer and transformer | tool | (Avsec et al. 2021) |
EWCE | Expression Weighted Cell-type Enrichment | tool | (Skene and Grant, 2016) |
ExPecto | ExPecto | tool | (Zhou et al. 2018) |
FINEMAP | FINEMAP | tool | (Benner et al., 2016) |
FitHiChIP | portmanteau of "Fit" and HiChIP | tool | (Bhattacharyya et al., 2019) |
H-MAGMA | Hi-C-coupled MAGMA | tool | (Sey et al., 2020) |
LDSC | Linkage Disequilibrium SCore regression | tool | (Finucane et al., 2015) |
MAGMA | Multi-marker Analysis of GenoMic Annotation | tool | (de Leeuw et al., 2015) |
MAGMA | Multi-marker Analysis of GenoMic Annotation | tool | |
MAPS | Model-based Analysis of PLAC-seq and HiChIP | tool | (Juric et al., 2019) |
MungeSumstats | MungeSumstats | tool | (Murphy and Skene, 2021) |
Footnotes
Data and Code Availability
All data and code used to produce the plots and meta-analyses described in this manuscript are publicly available via the dedicated GitHub repository: https://github.com/RajLabMSSM/PD_omics_review
Declarations of interest
None.
References
- Adamson B, Norman TM, Jost M, Cho MY, Nuñez JK, Chen Y, Villalta JE, Gilbert LA, Horlbeck MA, Hein MY, Pak RA, Gray AN, Gross CA, Dixit A, Parnas O, Regev A, Weissman JS, 2016. A Multiplexed Single-Cell CRISPR Screening Platform Enables Systematic Dissection of the Unfolded Protein Response. Cell 167, 1867–1882.e21. 10.1016/j.cell.2016.11.048 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Adli M, 2018. The CRISPR tool kit for genome editing and beyond. Nat. Commun 9, 1911. 10.1038/s41467-018-04252-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agarwal D, Sandor C, Volpato V, Caffrey TM, Monzón-Sandoval J, Bowden R, Alegre-Abarrategui J, Wade-Martins R, Webber C, 2020. A single-cell atlas of the human substantia nigra reveals cell-specific pathways associated with neurological disorders. Nat. Commun 11, 4183. 10.1038/s41467-020-17876-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aguirre M, Tanigawa Y, Venkataraman GR, Tibshirani R, Hastie T, Rivas MA, 2021. Polygenic risk modeling with latent trait-related genetic components. Eur. J. Hum. Genet 10.1038/s41431-021-00813-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aleksovski D, Miljkovic D, Bravi D, Antonini A, 2018. Disease progression in Parkinson subtypes: the PPMI dataset. Neurol. Sci 39, 1971–1976. 10.1007/s10072-018-3522-z [DOI] [PubMed] [Google Scholar]
- Alfradique-Dunham I, Al-Ouran R, von Coelln R, Blauwendraat C, Hill E, Luo L, Stillwell A, Young E, Kaw A, Tan M, Liao C, Hernandez D, Pihlstrom L, Grosset D, Shulman LM, Liu Z, Rouleau GA, Nalls M, Singleton AB, Morris H, Jankovic J, Shulman JM, 2021. Genome-Wide Association Study Meta-Analysis for Parkinson Disease Motor Subtypes. Neurology Genetics 7. 10.1212/NXG.0000000000000557 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Alyagor I, Berkun V, Keren-Shaul H, Marmor-Kollet N, David E, Mayseless O, Issman-Zecharya N, Amit I, Schuldiner O, 2018. Combining Developmental and Perturbation-Seq Uncovers Transcriptional Modules Orchestrating Neuronal Remodeling. Dev. Cell 47, 38–52.e6. 10.1016/j.devcel.2018.09.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Andersen MS, Bandres-Ciga S, Reynolds RH, Hardy J, Ryten M, Krohn L, Gan-Or Z, Holtman IR, International Parkinson’s Disease Genomics Consortium, Pihlstrøm, L., 2021. Heritability enrichment implicates microglia in Parkinson’s disease pathogenesis. Ann. Neurol 10.1002/ana.26032 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Annesley SJ, Lay ST, De Piazza SW, Sanislav O, Hammersley E, Allan CY, Francione LM, Bui MQ, Chen Z-P, Ngoei KRW, Tassone F, Kemp BE, Storey E, Evans A, Loesch DZ, Fisher PR, 2016. Immortalized Parkinson’s disease lymphocytes have enhanced mitochondrial respiratory activity. Dis. Model. Mech 9, 1295–1305. 10.1242/dmm.025684 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arai H, Furuya T, Mizuno Y, Mochizuki H, 2006. Inflammation and infection in Parkinson’s disease. Histol. Histopathol [DOI] [PubMed] [Google Scholar]
- Avsec Z, Agarwal V, Visentin D, Ledsam JR, 2021. Effective gene expression prediction from sequence by integrating long-range interactions. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Avsec Ž, Kreuzhuber R, Israeli J, Xu N, Cheng J, Shrikumar A, Banerjee A, Kim DS, Beier T, Urban L, Kundaje A, Stegle O, Gagneur J, 2019. The Kipoi repository accelerates community exchange and reuse of predictive models for genomics. Nat. Biotechnol 37, 592–600. 10.1038/s41587-019-0140-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Badanjak K, Fixemer S, Smajić S, Skupin A, Grünewald A, 2021. The Contribution of Microglia to Neuroinflammation in Parkinson’s Disease. International Journal of Molecular Sciences. 10.3390/ijms22094676 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Banati RB, Daniel SE, Blunt SB, 1998. Glial pathology but absence of apoptotic nigral neurons in long-standing Parkinson’s disease. Movement Disorders. 10.1002/mds.870130205 [DOI] [PubMed] [Google Scholar]
- Bandres-Ciga S, Diez-Fairen M, Kim JJ, Singleton AB, 2020. Genetics of Parkinson’s disease: An introspection of its journey towards precision medicine. Neurobiol. Dis 137, 104782. 10.1016/j.nbd.2020.104782 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bandres-Ciga S, Saez-Atienzar S, Bonet-Ponce L, Billingsley K, Vitale D, Blauwendraat C, Gibbs JR, Pihlstrøm L, Gan-Or Z, International Parkinson’s Disease Genomics Consortium (IPDGC), Cookson MR, Nalls MA, Singleton AB, 2019. The endocytic membrane trafficking pathway plays a major role in the risk of Parkinson’s disease. Mov. Disord. 34, 460–468. 10.1002/mds.27614 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bandres-Ciga S, Saez-Atienzar S, Kim JJ, Makarious MB, Faghri F, Diez-Fairen M, Iwaki H, Leonard H, Botia J, Ryten M, Hernandez D, Gibbs JR, Ding J, Gan-Or Z, Noyce A, Pihlstrom L, Torkamani A, Soltis AR, Dalgard CL, American Genome Center, Scholz SW, Traynor BJ, Ehrlich D, Scherzer CR, Bookman M, Cookson M, Blauwendraat C, Nalls MA, Singleton AB, International Parkinson Disease Genomics Consortium, 2020. Large-scale pathway specific polygenic risk and transcriptomic community network analysis identifies novel functional pathways in Parkinson disease. Acta Neuropathol. 10.1007/s00401-020-02181-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benner C, Havulinna AS, Järvelin M-R, Salomaa V, Ripatti S, Pirinen M, 2017. Prospects of Fine-Mapping Trait-Associated Genomic Regions by Using Summary Statistics from Genome-wide Association Studies. Am. J. Hum. Genet 101, 539–551. 10.1016/j.ajhg.2017.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Benner C, Spencer CCA, Havulinna AS, Salomaa V, Ripatti S, Pirinen M, 2016. FINEMAP: efficient variable selection using summary data from genome-wide association studies. Bioinformatics 32, 1493–1501. 10.1093/bioinformatics/btw018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhattacharyya S, Chandra V, Vijayanand P, Ay F, 2019. Identification of significant chromatin contacts from HiChIP data by FitHiChIP. Nat. Commun 10, 4221. 10.1038/s41467-019-11950-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Billingsley KJ, Barbosa IA, Bandrés-Ciga S, Quinn JP, Bubb VJ, Deshpande C, Botia JA, Reynolds RH, Zhang D, Simpson MA, Blauwendraat C, Gan-Or Z, Gibbs JR, Nalls MA, Singleton A, International Parkinson’s Disease Genomics Consortium (IPDGC), Ryten M, Koks S, 2019. Mitochondria function associated genes contribute to Parkinson’s Disease risk and later age at onset. NPJ Parkinsons Dis 5, 8. 10.1038/s41531-019-0080-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birgisdottir ÅB, Johansen T, 2020. Autophagy and endocytosis – interconnections and interdependencies. Journal of Cell Science. 10.1242/jcs.228114 [DOI] [PubMed] [Google Scholar]
- Blauwendraat C, Heilbron K, Vallerga CL, Bandres-Ciga S, Von Coelln R, Pihlstrøm L, Simón-Sánchez J, Schulte C, Sharma M, Krohn L, Others, 2019. Parkinson’s disease age at onset genome-wide association study: Defining heritability, genetic loci, and α-synuclein mechanisms. Mov. Disord 34, 866–875. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blauwendraat C, Nalls MA, Singleton AB, 2020. The genetic architecture of Parkinson’s disease. Lancet Neurol. 19, 170–178. 10.1016/S1474-4422(19)30287-X [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bocchi VD, Conforti P, Vezzoli E, Besusso D, Cappadona C, Lischetti T, Galimberti M, Ranzani V, Bonnal RJP, De Simone M, Rossetti G, He X, Kamimoto K, Espuny-Camacho I, Faedo A, Gervasoni F, Vuono R, Morris SA, Chen J, Felsenfeld D, Pavesi G, Barker RA, Pagani M, Cattaneo E, 2021. The coding and long noncoding single-cell atlas of the developing human fetal striatum. Science. 10.1126/science.abf5759 [DOI] [PubMed] [Google Scholar]
- Boix CA, James BT, Park YP, Meuleman W, Kellis M, 2021. Regulatory genomic circuitry of human disease loci by integrative epigenomics. Nature 590, 300–307. 10.1038/s41586-020-03145-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braak H, Del Tredici K, 2017. Neuropathological Staging of Brain Pathology in Sporadic Parkinson’s disease: Separating the Wheat from the Chaff. Journal of Parkinson’s Disease. 10.3233/jpd-179001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Braak H, Del Tredici K, Rüb U, de Vos RAI, Jansen Steur EN, Braak E, 2003. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiology of Aging. 10.1016/s0197-4580(02)00065-9 [DOI] [PubMed] [Google Scholar]
- Bressan E, Reed X, Bansal V, Hutchins E, Cobb MM, 2021. The Foundational data initiative for Parkinsons disease (FOUNDIN-PD): enabling efficient translation from genetic maps to mechanism. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brochard V, Combadière B, Prigent A, Laouar Y, Perrin A, Beray-Berthat V, Bonduelle O, Alvarez-Fischer D, Callebert J, Launay J-M, Duyckaerts C, Flavell RA, Hirsch EC, Hunot S, 2009. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J. Clin. Invest 119, 182–192. 10.1172/JCI36470 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Broekema RV, Bakker OB, Jonkers IH, 2020. A practical view of fine-mapping and gene prioritization in the post-genome-wide association era. Open Biol. 10, 190221. 10.1098/rsob.190221 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bryois J, Eating Disorders Working Group of the Psychiatric Genomics Consortium, Skene NG, Hansen TF, Kogelman LJA, Watson HJ, Liu Z, Brueggeman L, Breen G, Bulik CM, Arenas E, Hjerling-Leffler J, Sullivan PF, International Headache Genetics Consortium, 23andMe Research Team, 2020a. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nature Genetics. 10.1038/s41588-020-0610-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bryois J, Eating Disorders Working Group of the Psychiatric Genomics Consortium, Skene NG, Hansen TF, Kogelman LJA, Watson HJ, Liu Z, Brueggeman L, Breen G, Bulik CM, Arenas E, Hjerling-Leffler J, Sullivan PF, International Headache Genetics Consortium, 23andMe Research Team, 2020b. Genetic identification of cell types underlying brain complex traits yields insights into the etiology of Parkinson’s disease. Nature Genetics. 10.1038/s41588-020-0610-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burbulla LF, Song P, Mazzulli JR, Zampese E, Wong YC, Jeon S, Santos DP, Blanz J, Obermaier CD, Strojny C, Savas JN, Kiskinis E, Zhuang X, Krüger R, James Surmeier D, Krainc D, 2017. Dopamine oxidation mediates mitochondrial and lysosomal dysfunction in Parkinson’s disease. Science. 10.1126/science.aam9080 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bycroft C, Freeman C, Petkova D, Band G, Elliott LT, Sharp K, Motyer A, Vukcevic D, Delaneau O, O’Connell J, Cortes A, Welsh S, Young A, Effingham M, McVean G, Leslie S, Allen N, Donnelly P, Marchini J, 2018. The UK Biobank resource with deep phenotyping and genomic data. Nature 562, 203–209. 10.1038/s41586-018-0579-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cao J, O’Day DR, Pliner HA, Kingsley PD, Deng M, Daza RM, Zager MA, Aldinger KA, Blecher-Gonen R, Zhang F, Spielmann M, Palis J, Doherty D, Steemers FJ, Glass IA, Trapnell C, Shendure J, 2020. A human cell atlas of fetal gene expression. Science 370. 10.1126/science.aba7721 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carvalho-Silva D, Pierleoni A, Pignatelli M, Ong C, Fumis L, Karamanis N, Carmona M, Faulconbridge A, Hercules A, McAuley E, Miranda A, Peat G, Spitzer M, Barrett J, Hulcoop DG, Papa E, Koscielny G, Dunham I, 2019. Open Targets Platform: new developments and updates two years on. Nucleic Acids Res. 47, D1056–D1065. 10.1093/nar/gky1133 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chang D, Nalls MA, Hallgrímsdóttir IB, Hunkapiller J, van der Brug M, Cai F, International Parkinson’s Disease Genomics Consortium, 23andMe Research Team, Kerchner GA, Ayalon G, Bingol B, Sheng M, Hinds D, Behrens TW, Singleton AB, Bhangale TR, Graham RR, 2017. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nat. Genet 49, 1511–1516. 10.1038/ng.3955 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y-C, Farzadfard F, Gharaei N, Chen WCW, Cao J, Lu TK, 2017. Randomized CRISPR-Cas Transcriptional Perturbation Screening Reveals Protective Genes against Alpha-Synuclein Toxicity. Mol. Cell 68, 247–257.e5. 10.1016/j.molcel.2017.09.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chu K, Zhou X, Luo B-Y, 2012. Cytokine gene polymorphisms and Parkinson’s disease: a meta-analysis. Can. J. Neurol. Sci. 39, 58–64. 10.1017/s0317167100012695 [DOI] [PubMed] [Google Scholar]
- Coccia E, Ahfeldt T, 2021. Towards physiologically relevant human pluripotent stem cell (hPSC) models of Parkinson’s disease. Stem Cell Research & Therapy. 10.1186/s13287-021-02326-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coetzee SG, Pierce S, Brundin P, Brundin L, Hazelett DJ, Coetzee GA, 2016. Enrichment of risk SNPs in regulatory regions implicate diverse tissues in Parkinson’s disease etiology. Scientific Reports. 10.1038/srep30509 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cong L, Ran FA, Cox D, Lin S, Barretto R, Habib N, Hsu PD, Wu X, Jiang W, Marraffini LA, Zhang F, 2013. Multiplex genome engineering using CRISPR/Cas systems. Science 339, 819–823. 10.1126/science.1231143 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cook DA, Kannarkat GT, Cintron AF, Butkovich LM, Fraser KB, Chang J, Grigoryan N, Factor SA, West AB, Boss JM, Tansey MG, 2017. LRRK2 levels in immune cells are increased in Parkinson’s disease. NPJ Parkinsons Dis 3, 11. 10.1038/s41531-017-0010-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Corces MR, Shcherbina A, Kundu S, Gloudemans MJ, Frésard L, Granja JM, Louie BH, Eulalio T, Shams S, Bagdatli ST, Mumbach MR, Liu B, Montine KS, Greenleaf WJ, Kundaje A, Montgomery SB, Chang HY, Montine TJ, 2020. Single-cell epigenomic analyses implicate candidate causal variants at inherited risk loci for Alzheimer’s and Parkinson's diseases. Nat. Genet 52, 1158–1168. 10.1038/s41588-020-00721-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Damier P, Hirsch EC, Agid Y, Graybiel AM, 1999. The substantia nigra of the human brain. II. Patterns of loss of dopamine-containing neurons in Parkinson’s disease. Brain 122 ( Pt 8), 1437–1448. 10.1093/brain/122.8.1437 [DOI] [PubMed] [Google Scholar]
- de Leeuw CA, Mooij JM, Heskes T, Posthuma D, 2015. MAGMA: generalized gene-set analysis of GWAS data. PLoS Comput. Biol 11, e1004219. 10.1371/journal.pcbi.1004219 [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Paiva Lopes K, Snijders GJL, Humphrey J, Allan A, 2020. Atlas of genetic effects in human microglia transcriptome across brain regions, aging and disease pathologies. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Witte L, Frucht S, Saunders-Pullman R, Raj T, 2020. Discordant transcriptional signatures of mitochondrial genes in Parkinson’s disease human myeloid cells. bioRxiv. [Google Scholar]
- Dey KK, van de Geijn B, Kim SS, Hormozdiari F, Kelley DR, Price AL, 2020. Evaluating the informativeness of deep learning annotations for human complex diseases. Nat. Commun 11, 4703. 10.1038/s41467-020-18515-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dijkstra AA, Ingrassia A, de Menezes RX, van Kesteren RE, Rozemuller AJM, Heutink P, van de Berg WDJ, 2015. Evidence for Immune Response, Axonal Dysfunction and Reduced Endocytosis in the Substantia Nigra in Early Stage Parkinson’s Disease. PLOS ONE. 10.1371/journal.pone.0128651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dixit A, Parnas O, Li B, Chen J, Fulco CP, Jerby-Arnon L, Marjanovic ND, Dionne D, Burks T, Raychowdhury R, Adamson B, Norman TM, Lander ES, Weissman JS, Friedman N, Regev A, 2016. Perturb-Seq: Dissecting Molecular Circuits with Scalable Single-Cell RNA Profiling of Pooled Genetic Screens. Cell 167, 1853–1866.e17. 10.1016/j.cell.2016.11.038 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dobbs RJ, Charlett A, Purkiss AG, Dobbs SM, Weller C, Peterson DW, 1999. Association of circulating TNF-α and IL-6 with ageing and parkinsonism. Acta Neurol. Scand 100, 34–41. [DOI] [PubMed] [Google Scholar]
- Domcke S, Hill AJ, Daza RM, Cao J, O’Day DR, Pliner HA, Aldinger KA, Pokholok D, Zhang F, Milbank JH, Zager MA, Glass IA, Steemers FJ, Doherty D, Trapnell C, Cusanovich DA, Shendure J, 2020. A human cell atlas of fetal chromatin accessibility. Science 370. 10.1126/science.aba7612 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dorsey ER, Constantinescu R, Thompson JP, Biglan KM, Holloway RG, Kieburtz K, Marshall FJ, Ravina BM, Schifitto G, Siderowf A, Tanner CM, 2007. Projected number of people with Parkinson disease in the most populous nations, 2005 through 2030. Neurology 68, 384–386. 10.1212/01.wnl.0000247740.47667.03 [DOI] [PubMed] [Google Scholar]
- Elsworth B, Lyon M, Alexander T, Liu Y, Matthews P, Hallett J, Bates P, Palmer T, Haberland V, Smith GD, Zheng J, Haycock P, Gaunt TR, Hemani G, 2020. The MRC IEU OpenGWAS data infrastructure. bioRxiv. 10.1101/2020.08.10.244293 [DOI] [Google Scholar]
- Eraslan G, Avsec Ž, Gagneur J, Theis FJ, 2019. Deep learning: new computational modelling techniques for genomics. Nat. Rev. Genet 20, 389–403. 10.1038/s41576-019-0122-6 [DOI] [PubMed] [Google Scholar]
- Faa G, Marcialis MA, Ravarino A, Piras M, Pintus MC, Fanos V, 2014. Fetal programming of the human brain: is there a link with insurgence of neurodegenerative disorders in adulthood? Curr. Med. Chem 21, 3854–3876. 10.2174/0929867321666140601163658 [DOI] [PubMed] [Google Scholar]
- Fairfax BP, Humburg P, Makino S, Naranbhai V, Wong D, Lau E, Jostins L, Plant K, Andrews R, McGee C, Knight JC, 2014. Innate immune activity conditions the effect of regulatory variants upon monocyte gene expression. Science 343, 1246949. 10.1126/science.1246949 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Falconer DS, 1996. Introduction to quantitative genetics. Pearson Education India. [Google Scholar]
- Fang R, Yu M, Li G, Chee S, Liu T, Schmitt AD, Ren B, 2016. Mapping of long-range chromatin interactions by proximity ligation-assisted ChIP-seq. Cell Res. 26, 1345–1348. 10.1038/cr.2016.137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farh KK-H, Marson A, Zhu J, Kleinewietfeld M, Housley WJ, Beik S, Shoresh N, Whitton H, Ryan RJH, Shishkin AA, Hatan M, Carrasco-Alfonso MJ, Mayer D, Luckey CJ, Patsopoulos NA, De Jager PL, Kuchroo VK, Epstein CB, Daly MJ, Hafler DA, Bernstein BE, 2015. Genetic and epigenetic fine mapping of causal autoimmune disease variants. Nature 518, 337–343. 10.1038/nature13835 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fearnley JM, Lees AJ, 1991. Ageing and Parkinson’s disease: substantia nigra regional selectivity. Brain 114 ( Pt 5), 2283–2301. 10.1093/brain/114.5.2283 [DOI] [PubMed] [Google Scholar]
- Fernández-Santiago R, Carballo-Carbajal I, Castellano G, Torrent R, Richaud Y, Sánchez-Danés A, Vilarrasa-Blasi R, Sánchez-Pla A, Mosquera JL, Soriano J, López-Barneo J, Canals JM, Alberch J, Raya Á, Vila M, Consiglio A, Martín-Subero JI, Ezquerra M, Tolosa E, 2015. Aberrant epigenome in iPSC-derived dopaminergic neurons from Parkinson’s disease patients. EMBO Mol. Med 7, 1529–1546. 10.15252/emmm.201505439 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernández-Santiago R, Merkel A, Castellano G, Heath S, Raya Á, Tolosa E, Martí M-J, Consiglio A, Ezquerra M, 2019. Whole-genome DNA hyper-methylation in iPSC-derived dopaminergic neurons from Parkinson’s disease patients. Clin. Epigenetics 11, 108. 10.1186/s13148-019-0701-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fernando MB, Ahfeldt T, Brennand KJ, 2020. Modeling the complex genetic architectures of brain disease. Nat. Genet 52, 363–369. 10.1038/s41588-020-0596-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Finucane HK, Bulik-Sullivan B, Gusev A, Trynka G, Reshef Y, Loh P-R, Anttila V, Xu H, Zang C, Farh K, Ripke S, Day FR, ReproGen Consortium, Schizophrenia Working Group of the Psychiatric Genomics Consortium, RACI Consortium, Purcell S, Stahl E, Lindstrom S, Perry JRB, Okada Y, Raychaudhuri S, Daly MJ, Patterson N, Neale BM, Price AL, 2015. Partitioning heritability by functional annotation using genome-wide association summary statistics. Nat. Genet 47, 1228–1235. 10.1038/ng.3404 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fong H, Wang C, Knoferle J, Walker D, Balestra ME, Tong LM, Leung L, Ring KL, Seeley WW, Karydas A, Kshirsagar MA, Boxer AL, Kosik KS, Miller BL, Huang Y, 2013. Genetic correction of tauopathy phenotypes in neurons derived from human induced pluripotent stem cells. Stem Cell Reports 1, 226–234. 10.1016/j.stemcr.2013.08.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fromer M, Roussos P, Sieberts SK, Johnson JS, Kavanagh DH, Perumal TM, Ruderfer DM, Oh EC, Topol A, Shah HR, Klei LL, Kramer R, Pinto D, Gümüş ZH, Cicek AE, Dang KK, Browne A, Lu C, Xie L, Readhead B, Stahl EA, Xiao J, Parvizi M, Hamamsy T, Fullard JF, Wang Y-C, Mahajan MC, Derry JMJ, Dudley JT, Hemby SE, Logsdon BA, Talbot K, Raj T, Bennett DA, De Jager PL, Zhu J, Zhang B, Sullivan PF, Chess A, Purcell SM, Shinobu LA, Mangravite LM, Toyoshiba H, Gur RE, Hahn C-G, Lewis DA, Haroutunian V, Peters MA, Lipska BK, Buxbaum JD, Schadt EE, Hirai K, Roeder K, Brennand KJ, Katsanis N, Domenici E, Devlin B, Sklar P, 2016. Gene expression elucidates functional impact of polygenic risk for schizophrenia. Nat. Neurosci 19, 1442–1453. 10.1038/nn.4399 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fulco CP, Nasser J, Jones TR, Munson G, Bergman DT, Subramanian V, Grossman SR, Anyoha R, Doughty BR, Patwardhan TA, Nguyen TH, Kane M, Perez EM, Durand NC, Lareau CA, Stamenova EK, Aiden EL, Lander ES, Engreitz JM, 2019. Activity-by-contact model of enhancer-promoter regulation from thousands of CRISPR perturbations. Nat. Genet 51, 1664–1669. 10.1038/s41588-019-0538-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gagliano SA, Pouget JG, Hardy J, Knight J, Barnes MR, Ryten M, Weale ME, 2016. Genomics implicates adaptive and innate immunity in Alzheimer’s and Parkinson’s diseases. Annals of Clinical and Translational Neurology. 10.1002/acn3.369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Galet B, Cheval H, Ravassard P, 2020. Patient-Derived Midbrain Organoids to Explore the Molecular Basis of Parkinson’s Disease. Front. Neurol 11, 1005. 10.3389/fneur.2020.01005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K, Carroll RJ, Eyler AE, Denny JC, GTEx Consortium, Nicolae DL, Cox NJ, Im HK, 2015. A gene-based association method for mapping traits using reference transcriptome data. Nat. Genet 47, 1091–1098. 10.1038/ng.3367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garcia-Ruiz PJ, Espay AJ, 2017. Parkinson Disease: An Evolutionary Perspective. Front. Neurol 8, 157. 10.3389/fneur.2017.00157 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garretti F, Agalliu D, Lindestam Arlehamn CS, Sette A, Sulzer D, 2019. Autoimmunity in Parkinson’s Disease: The Role of α-Synuclein-Specific T Cells. Front. Immunol 10, 303. 10.3389/fimmu.2019.00303 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garrido-Martín D, Borsari B, Calvo M, Reverter F, Guigó R, 2021. Identification and analysis of splicing quantitative trait loci across multiple tissues in the human genome. Nat. Commun 12, 727. 10.1038/s41467-020-20578-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gerhard A, Pavese N, Hotton G, Turkheimer F, Es M, Hammers A, Eggert K, Oertel W, Banati RB, Brooks DJ, 2006. In vivo imaging of microglial activation with [11C](R)-PK11195 PET in idiopathic Parkinson’s disease. Neurobiol. Dis 21, 404–412. 10.1016/j.nbd.2005.08.002 [DOI] [PubMed] [Google Scholar]
- Giambartolomei C, Vukcevic D, Schadt EE, Franke L, Hingorani AD, Wallace C, Plagnol V, 2014. Bayesian test for colocalisation between pairs of genetic association studies using summary statistics. PLoS Genet. 10, e1004383. 10.1371/journal.pgen.1004383 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gosselin D, Skola D, Coufal NG, Holtman IR, Schlachetzki JCM, Sajti E, Jaeger BN, O’Connor C, Fitzpatrick C, Pasillas MP, Pena M, Adair A, Gonda DD, Levy ML, Ransohoff RM, Gage FH, Glass CK, 2017. An environment-dependent transcriptional network specifies human microglia identity. Science 356. 10.1126/science.aal3222 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Greene CS, Krishnan A, Wong AK, Ricciotti E, Zelaya RA, Himmelstein DS, Zhang R, Hartmann BM, Zaslavsky E, Sealfon SC, Chasman DI, FitzGerald GA, Dolinski K, Grosser T, Troyanskaya OG, 2015. Understanding multicellular function and disease with human tissue-specific networks. Nat. Genet 47, 569–576. 10.1038/ng.3259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grenn FP, Kim JJ, Makarious MB, Iwaki H, Illarionova A, Brolin K, Kluss JH, Schumacher-Schuh AF, Leonard H, Faghri F, Billingsley K, Krohn L, Hall A, Diez-Fairen M, Periñán MT, Foo JN, Sandor C, Webber C, Fiske BK, Gibbs JR, Nalls MA, Singleton AB, Bandres-Ciga S, Reed X, Blauwendraat C, International Parkinson’s Disease Genomics Consortium (IPDGC), 2020. The Parkinson’s Disease Genome-Wide Association Study Locus Browser. Mov. Disord 10.1002/mds.28197 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grozdanov V, Bliederhaeuser C, Ruf WP, Roth V, Fundel-Clemens K, Zondler L, Brenner D, Martin-Villalba A, Hengerer B, Kassubek J, Ludolph AC, Weishaupt JH, Danzer KM, 2014. Inflammatory dysregulation of blood monocytes in Parkinson’s disease patients. Acta Neuropathol. 128, 651–663. 10.1007/s00401-014-1345-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grozdanov V, Bousset L, Hoffmeister M, Bliederhaeuser C, Meier C, Madiona K, Pieri L, Kiechle M, McLean PJ, Kassubek J, Behrends C, Ludolph AC, Weishaupt JH, Melki R, Danzer KM, 2019. Increased Immune Activation by Pathologic α-Synuclein in Parkinson’s Disease. Ann. Neurol 86, 593–606. 10.1002/ana.25557 [DOI] [PubMed] [Google Scholar]
- Consortium GTEx, 2015. Human genomics. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science 348, 648–660. 10.1126/science.1262110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gusev A, Ko A, Shi H, Bhatia G, Chung W, Penninx BWJH, Jansen R, de Geus EJC, Boomsma DI, Wright FA, Sullivan PF, Nikkola E, Alvarez M, Civelek M, Lusis AJ, Lehtimäki T, Raitoharju E, Kähönen M, Seppälä I, Raitakari OT, Kuusisto J, Laakso M, Price AL, Pajukanta P, Pasaniuc B, 2016. Integrative approaches for large-scale transcriptome-wide association studies. Nat. Genet 48, 245–252. 10.1038/ng.3506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gusev A, Mancuso N, Won H, Kousi M, Finucane HK, Reshef Y, Song L, Safi A, Schizophrenia Working Group of the Psychiatric Genomics Consortium, McCarroll S, Neale BM, Ophoff RA, O’Donovan MC, Crawford GE, Geschwind DH, Katsanis N, Sullivan PF, Pasaniuc B, Price AL, 2018. Transcriptome-wide association study of schizophrenia and chromatin activity yields mechanistic disease insights. Nat. Genet 50, 538–548. 10.1038/s41588-018-0092-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hamza TH, Zabetian CP, Tenesa A, Laederach A, Montimurro J, Yearout D, Kay DM, Doheny KF, Paschall J, Pugh E, Kusel VI, Collura R, Roberts J, Griffith A, Samii A, Scott WK, Nutt J, Factor SA, Payami H, 2010. Common genetic variation in the HLA region is associated with late-onset sporadic Parkinson’s disease. Nat. Genet 42, 781–785. 10.1038/ng.642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Heng TSP, Painter MW, Immunological Genome Project Consortium, 2008. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol 9, 1091–1094. 10.1038/ni1008-1091 [DOI] [PubMed] [Google Scholar]
- Ho MS, 2019. Microglia in Parkinson’s Disease. Adv. Exp. Med. Biol 1175, 335–353. 10.1007/978-981-13-9913-8_13 [DOI] [PubMed] [Google Scholar]
- Hutchinson A, Asimit J, Wallace C, 2020. Fine-mapping genetic associations. Hum. Mol. Genet 29, R81–R88. 10.1093/hmg/ddaa148 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Imamura K, Hishikawa N, Sawada M, Nagatsu T, Yoshida M, Hashizume Y, 2003. Distribution of major histocompatibility complex class II-positive microglia and cytokine profile of Parkinson’s disease brains. Acta Neuropathol. 106, 518–526. 10.1007/s00401-003-0766-2 [DOI] [PubMed] [Google Scholar]
- Iwaki H, Leonard HL, Makarious MB, Bookman M, Landin B, Vismer D, Casey B, Gibbs JR, Hernandez DG, Blauwendraat C, Others, 2020. Accelerating Medicines Partnership: Parkinson’s Disease. Genetic Resource. medRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jagmag SA, Tripathi N, Shukla SD, Maiti S, Khurana S, 2015. Evaluation of Models of Parkinson’s Disease. Front. Neurosci 9, 503. 10.3389/fnins.2015.00503 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jansen IE, International Parkinson’s Disease Genetics Consortium (IPGDC), Ye H, Heetveld S, Lechler MC, Michels H, Seinstra RI, Lubbe SJ, Drouet V, Lesage S, Majounie E, Raphael Gibbs J, Nalls MA, Ryten M, Botia JA, Vandrovcova J, Simon-Sanchez J, Castillo-Lizardo M, Rizzu P, Blauwendraat C, Chouhan AK, Li Y, Yogi P, Amin N, van Duijn CM, Morris HR, Brice A, Singleton AB, David DC, Nollen EA, Jain S, Shulman JM, Heutink P, 2017. Discovery and functional prioritization of Parkinson’s disease candidate genes from large-scale whole exome sequencing. Genome Biology. 10.1186/s13059-017-1147-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jin X, Simmons SK, Guo A, Shetty AS, Ko M, Nguyen L, Jokhi V, Robinson E, Oyler P, Curry N, Deangeli G, Lodato S, Levin JZ, Regev A, Zhang F, Arlotta P, 2020. In vivo Perturb-Seq reveals neuronal and glial abnormalities associated with autism risk genes. Science 370. 10.1126/science.aaz6063 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Juric I, Yu M, Abnousi A, Raviram R, Fang R, Zhao Y, Zhang Y, Qiu Y, Yang Y, Li Y, Ren B, Hu M, 2019. MAPS: Model-based analysis of long-range chromatin interactions from PLAC-seq and HiChIP experiments. PLoS Comput. Biol. 15, e1006982. 10.1371/journal.pcbi.1006982 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalia LV, Lang AE, 2015. Parkinson’s disease. Lancet 386, 896–912. 10.1016/S0140-6736(14)61393-3 [DOI] [PubMed] [Google Scholar]
- Kamath T, Abdulraouf A, Burris SJ, Gazestani V, Nadaf N, Vanderburg C, Macosko EZ, 2021. A molecular census of midbrain dopaminergic neurons in Parkinson’s disease. bioRxiv. 10.1101/2021.06.16.448661 [DOI] [Google Scholar]
- Kam T-I, Hinkle JT, Dawson TM, Dawson VL, 2020. Microglia and astrocyte dysfunction in parkinson’s disease. Neurobiol. Dis 144, 105028. 10.1016/j.nbd.2020.105028 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kannarkat GT, Boss JM, Tansey MG, 2013. The role of innate and adaptive immunity in Parkinson’s disease. J. Parkinsons. Dis 3, 493–514. 10.3233/JPD-130250 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kearsey MJ, 1998. The principles of QTL analysis (a minimal mathematics approach). J. Exp. Bot 49, 1619–1623. 10.1093/jxb/49.327.1619 [DOI] [Google Scholar]
- Keller MF, Saad M, Bras J, Bettella F, 2012. Using genome-wide complex trait analysis to quantify “missing heritability”in Parkinson’s disease. Hum. Mol. Genet [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kelley DR, Reshef YA, Bileschi M, Belanger D, McLean CY, Snoek J, 2018. Sequential regulatory activity prediction across chromosomes with convolutional neural networks. Genome Res. 28, 739–750. 10.1101/gr.227819.117 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kerimov N, Hayhurst JD, Manning JR, Walter P, Kolberg L, Peikova K, Samoviča M, Burdett T, Jupp S, Parkinson H, Papatheodorou I, Zerbino DR, Alasoo K, 2020. eQTL Catalogue: a compendium of uniformly processed human gene expression and splicing QTLs. Cold Spring Harbor Laboratory. 10.1101/2020.01.29.924266 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kichaev G, Roytman M, Johnson R, Eskin E, Lindström S, Kraft P, Pasaniuc B, 2017. Improved methods for multi-trait fine mapping of pleiotropic risk loci. Bioinformatics 33, 248–255. 10.1093/bioinformatics/btw615 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim H, Park HJ, Choi H, Chang Y, Park H, Shin J, Kim J, Lengner CJ, Lee YK, Kim J, 2019. Modeling G2019S-LRRK2 Sporadic Parkinson’s Disease in 3D Midbrain Organoids. Stem Cell Reports 12, 518–531. 10.1016/j.stemcr.2019.01.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kim SD, Allen NE, Canning CG, Fung VSC, 2018. Parkinson disease. Handb. Clin. Neurol 159, 173–193. 10.1016/B978-0-444-63916-5.00011-2 [DOI] [PubMed] [Google Scholar]
- Kopp W, Monti R, Tamburrini A, Ohler U, Akalin A, 2020. Deep learning for genomics using Janggu. Nat. Commun 11, 3488. 10.1038/s41467-020-17155-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kovacs GG, Adle-Biassette H, Milenkovic I, Cipriani S, van Scheppingen J, Aronica E, 2014. Linking pathways in the developing and aging brain with neurodegeneration. Neuroscience 269, 152–172. 10.1016/j.neuroscience.2014.03.045 [DOI] [PubMed] [Google Scholar]
- Lachmann A, Schilder BM, Wojciechowicz ML, Torre D, Kuleshov MV, Keenan AB, Ma’ayan A, 2019. Geneshot: search engine for ranking genes from arbitrary text queries. Nucleic Acids Res. 47, W571–W577. 10.1093/nar/gkz393 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langston RG, Beilina A, Reed X, Singleton AB, 2021a. Association of a Common Genetic Variant with Parkinson’s Disease is Propagated through Microglia. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Langston RG, Beilina A, Reed X, Singleton AB, Blauwendraat C, Gibbs JR, Cookson MR, 2021b. Association of a Common Genetic Variant with Parkinson’s Disease is Propagated through Microglia. bioRxiv. 10.1101/2021.01.15.426824 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lesage S, Brice A, 2009. Parkinson’s disease: from monogenic forms to genetic susceptibility factors. Hum. Mol. Genet. 18, R48–59. 10.1093/hmg/ddp012 [DOI] [PubMed] [Google Scholar]
- Lesnick TG, Papapetropoulos S, Mash DC, Ffrench-Mullen J, Shehadeh L, de Andrade M, Henley JR, Rocca WA, Ahlskog JE, Maraganore DM, 2007. A genomic pathway approach to a complex disease: axon guidance and Parkinson disease. PLoS Genet. 3, e98. 10.1371/journal.pgen.0030098 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li B, Zhao G, Li K, Wang Z, Fang Z, Wang X, Luo T, Zhang Y, Wang Y, Chen Q, Huang Y, Dong L, Guo J, Tang B, Li J, 2021. Characterizing the Expression Patterns of Parkinson’s Disease Associated Genes. Frontiers in Neuroscience. 10.3389/fnins.2021.629156 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lieberman-Aiden E, van Berkum NL, Williams L, Imakaev M, Ragoczy T, Telling A, Amit I, Lajoie BR, Sabo PJ, Dorschner MO, Sandstrom R, Bernstein B, Bender MA, Groudine M, Gnirke A, Stamatoyannopoulos J, Mirny LA, Lander ES, Dekker J, 2009. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293. 10.1126/science.1181369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lill CM, Roehr JT, McQueen MB, Kavvoura FK, Bagade S, Schjeide B-MM, Schjeide LM, Meissner E, Zauft U, Allen NC, Liu T, Schilling M, Anderson KJ, Beecham G, Berg D, Biernacka JM, Brice A, DeStefano AL, Do CB, Eriksson N, Factor SA, Farrer MJ, Foroud T, Gasser T, Hamza T, Hardy JA, Heutink P, Hill-Burns EM, Klein C, Latourelle JC, Maraganore DM, Martin ER, Martinez M, Myers RH, Nalls MA, Pankratz N, Payami H, Satake W, Scott WK, Sharma M, Singleton AB, Stefansson K, Toda T, Tung JY, Vance J, Wood NW, Zabetian CP, 23andMe Genetic Epidemiology of Parkinson’s Disease Consortium, International Parkinson’s Disease Genomics Consortium, Parkinson’s Disease GWAS Consortium, Wellcome Trust Case Control Consortium 2), Young P, Tanzi RE, Khoury MJ, Zipp F, Lehrach H, Ioannidis JPA, Bertram L, 2012. Comprehensive research synopsis and systematic meta-analyses in Parkinson’s disease genetics: The PDGene database. PLoS Genet. 8, e1002548. 10.1371/journal.pgen.1002548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu Y, Elsworth B, Erola P, Haberland V, Hemani G, Lyon M, Zheng J, Lloyd O, Vabistsevits M, Gaunt TR, 2020. EpiGraphDB: a database and data mining platform for health data science. Bioinformatics. 10.1093/bioinformatics/btaa961 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Kim Y, Tsang EK, Davis JR, Damani FN, Chiang C, Hess GT, Zappala Z, Strober BJ, Scott AJ, Li A, Ganna A, Bassik MC, Merker JD, GTEx Consortium, Laboratory, Data Analysis &Coordinating Center (LDACC)—Analysis Working Group, Statistical Methods groups—Analysis Working Group, Enhancing GTEx (eGTEx) groups, NIH Common Fund, NIH/NCI, NIH/NHGRI, NIH/NIMH, NIH/NIDA, Biospecimen Collection Source Site—NDRI, Biospecimen Collection Source Site—RPCI, Biospecimen Core Resource—VARI, Brain Bank Repository—University of Miami Brain Endowment Bank, Leidos Biomedical—Project Management, ELSI Study, Genome Browser Data Integration &Visualization—EBI, Genome Browser Data Integration &Visualization—UCSC Genomics Institute, University of California Santa Cruz, Hall IM, Battle A, Montgomery SB, 2017. The impact of rare variation on gene expression across tissues. Nature 550, 239–243. 10.1038/nature24267 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li X, Sundquist J, Sundquist K, 2012. Subsequent risks of Parkinson disease in patients with autoimmune and related disorders: a nationwide epidemiological study from Sweden. Neurodegener. Dis 10, 277–284. 10.1159/000333222 [DOI] [PubMed] [Google Scholar]
- Li YI, Wong G, Humphrey J, Raj T, 2019. Prioritizing Parkinson’s disease genes using population-scale transcriptomic data. Nat. Commun. 10, 994. 10.1038/s41467-019-08912-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Makarious MB, Leonard HL, Vitale D, Iwaki H, Saffo D, Sargent L, Dadu A, Castaño ES, Carter JF, Maleknia M, Botia JA, Blauwendraat C, Campbell RH, Hashemi SH, Singleton AB, Nalls MA, Faghri F, 2021. GenoML: Automated Machine Learning for Genomics. arXiv [cs.LG]. [Google Scholar]
- Marrone L, Bus C, Schöndorf D, Fitzgerald JC, Kübler M, Schmid B, Reinhardt P, Reinhardt L, Deleidi M, Levin T, Meixner A, Klink B, Glatza M, Gloeckner CJ, Gasser T, Sterneckert J, 2018. Generation of iPSCs carrying a common LRRK2 risk allele for in vitro modeling of idiopathic Parkinson’s disease. PLoS One 13, e0192497. 10.1371/journal.pone.0192497 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McGeer PL, Itagaki S, Boyes BE, McGeer EG, 1988. Reactive microglia are positive for HLA-DR in the substantia nigra of Parkinson’s and Alzheimer's disease brains. Neurology 38, 1285–1285. 10.1212/WNL.38.8.1285 [DOI] [PubMed] [Google Scholar]
- McGeer PL, McGeer EG, 2004. Inflammation and neurodegeneration in Parkinson’s disease. Parkinsonism Relat. Disord 10 Suppl 1, S3–7. 10.1016/j.parkreldis.2004.01.005 [DOI] [PubMed] [Google Scholar]
- McInnes G, Tanigawa Y, DeBoever C, Lavertu A, Olivieri JE, Aguirre M, Rivas MA, 2019. Global Biobank Engine: enabling genotype-phenotype browsing for biobank summary statistics. Bioinformatics 35, 2495–2497. 10.1093/bioinformatics/bty999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- McInnes L, Healy J, Melville J, 2018. UMAP: Uniform Manifold Approximation and Projection for Dimension Reduction. arXiv [stat.ML]. [Google Scholar]
- Mirza B, Hadberg H, Thomsen P, Moos T, 2000. The absence of reactive astrocytosis is indicative of a unique inflammatory process in Parkinson’s disease. Neuroscience 95, 425–432. 10.1016/s0306-4522(99)00455-8 [DOI] [PubMed] [Google Scholar]
- Mizuno Y, Ohta S, Tanaka M, Takamiya S, Suzuki K, Sato T, Oya H, Ozawa T, Kagawa Y, 1989. Deficiencies in Complex I subunits of the respiratory chain in Parkinson’s disease. Biochemical and Biophysical Research Communications. 10.1016/0006-291x(89)91141-8 [DOI] [PubMed] [Google Scholar]
- Mogi M, Harada M, Kondo T, Narabayashi H, Riederer P, Nagatsu T, 1995. Transforming growth factor-β1 levels are elevated in the striatum and in ventricular cerebrospinal fluid in Parkinson’s disease. Neurosci. Lett 193, 129–132. 10.1016/0304-3940(95)11686-Q [DOI] [PubMed] [Google Scholar]
- Mogi M, Harada M, Riederer P, Narabayashi H, Fujita K, Nagatsu T, 1994. Tumor necrosis factor-α (TNF-α) increases both in the brain and in the cerebrospinal fluid from parkinsonian patients. Neurosci. Lett 165, 208–210. 10.1016/0304-3940(94)90746-3 [DOI] [PubMed] [Google Scholar]
- Moran LB, Duke DC, Deprez M, Dexter DT, Pearce RKB, Graeber MB, 2006. Whole genome expression profiling of the medial and lateral substantia nigra in Parkinson’s disease. Neurogenetics 7, 1–11. 10.1007/s10048-005-0020-2 [DOI] [PubMed] [Google Scholar]
- Mortiboys H, Johansen KK, Aasly JO, Bandmann O, 2010. Mitochondrial impairment in patients with Parkinson disease with the G2019S mutation in LRRK2. Neurology 75, 2017–2020. 10.1212/WNL.0b013e3181ff9685 [DOI] [PubMed] [Google Scholar]
- Mulvey B, Lagunas T Jr, Dougherty JD, 2021. Massively Parallel Reporter Assays: Defining Functional Psychiatric Genetic Variants Across Biological Contexts. Biol. Psychiatry 89, 76–89. 10.1016/j.biopsych.2020.06.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mumbach MR, Rubin AJ, Flynn RA, Dai C, Khavari PA, Greenleaf WJ, Chang HY, 2016. HiChIP: efficient and sensitive analysis of protein-directed genome architecture. Nat. Methods 13, 919–922. 10.1038/nmeth.3999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mumbach MR, Satpathy AT, Boyle EA, Dai C, Gowen BG, Cho SW, Nguyen ML, Rubin AJ, Granja JM, Kazane KR, Wei Y, Nguyen T, Greenside PG, Corces MR, Tycko J, Simeonov DR, Suliman N, Li R, Xu J, Flynn RA, Kundaje A, Khavari PA, Marson A, Corn JE, Quertermous T, Greenleaf WJ, Chang HY, 2017. Enhancer connectome in primary human cells identifies target genes of disease-associated DNA elements. Nat. Genet 49, 1602–1612. 10.1038/ng.3963 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murphy AE, Skene NG, 2021. MungeSumstats: A Bioconductor package for the standardisation and quality control of many GWAS summary statistics. bioRxiv. 10.1101/2021.06.21.449239 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, Tan M, Kia DA, Noyce AJ, Xue A, Bras J, Young E, von Coelln R, Simón-Sánchez J, Schulte C, Sharma M, Krohn L, Pihlstrøm L, Siitonen A, Iwaki H, Leonard H, Faghri F, Gibbs JR, Hernandez DG, Scholz SW, Botia JA, Martinez M, Corvol J-C, Lesage S, Jankovic J, Shulman LM, Sutherland M, Tienari P, Majamaa K, Toft M, Andreassen OA, Bangale T, Brice A, Yang J, Gan-Or Z, Gasser T, Heutink P, Shulman JM, Wood NW, Hinds DA, Hardy JA, Morris HR, Gratten J, Visscher PM, Graham RR, Singleton AB, 23andMe Research Team, System Genomics of Parkinson’s Disease Consortium, International Parkinson’s Disease Genomics Consortium, 2019a. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 18, 1091–1102. 10.1016/S1474-4422(19)30320-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, Tan M, Kia DA, Noyce AJ, Xue A, Bras J, Young E, von Coelln R, Simón-Sánchez J, Schulte C, Sharma M, Krohn L, Pihlstrøm L, Siitonen A, Iwaki H, Leonard H, Faghri F, Gibbs JR, Hernandez DG, Scholz SW, Botia JA, Martinez M, Corvol J-C, Lesage S, Jankovic J, Shulman LM, Sutherland M, Tienari P, Majamaa K, Toft M, Andreassen OA, Bangale T, Brice A, Yang J, Gan-Or Z, Gasser T, Heutink P, Shulman JM, Wood NW, Hinds DA, Hardy JA, Morris HR, Gratten J, Visscher PM, Graham RR, Singleton AB, 23andMe Research Team, System Genomics of Parkinson’s Disease Consortium, International Parkinson’s Disease Genomics Consortium, 2019b. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 18, 1091–1102. 10.1016/S1474-4422(19)30320-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, Tan M, Kia DA, Noyce AJ, Xue A, Bras J, Young E, von Coelln R, Simón-Sánchez J, Schulte C, Sharma M, Krohn L, Pihlstrøm L, Siitonen A, Iwaki H, Leonard H, Faghri F, Gibbs JR, Hernandez DG, Scholz SW, Botia JA, Martinez M, Corvol J-C, Lesage S, Jankovic J, Shulman LM, Sutherland M, Tienari P, Majamaa K, Toft M, Andreassen OA, Bangale T, Brice A, Yang J, Gan-Or Z, Gasser T, Heutink P, Shulman JM, Wood NW, Hinds DA, Hardy JA, Morris HR, Gratten J, Visscher PM, Graham RR, Singleton AB, 23andMe Research Team, System Genomics of Parkinson’s Disease Consortium, International Parkinson’s Disease Genomics Consortium, 2019c. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 18, 1091–1102. 10.1016/S1474-4422(19)30320-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nalls MA, Blauwendraat C, Vallerga CL, Heilbron K, Bandres-Ciga S, Chang D, Tan M, Kia DA, Noyce AJ, Xue A, Bras J, Young E, von Coelln R, Simón-Sánchez J, Schulte C, Sharma M, Krohn L, Pihlstrøm L, Siitonen A, Iwaki H, Leonard H, Faghri F, Gibbs JR, Hernandez DG, Scholz SW, Botia JA, Martinez M, Corvol J-C, Lesage S, Jankovic J, Shulman LM, Sutherland M, Tienari P, Majamaa K, Toft M, Andreassen OA, Bangale T, Brice A, Yang J, Gan-Or Z, Gasser T, Heutink P, Shulman JM, Wood NW, Hinds DA, Hardy JA, Morris HR, Gratten J, Visscher PM, Graham RR, Singleton AB, 23andMe Research Team, System Genomics of Parkinson’s Disease Consortium, International Parkinson’s Disease Genomics Consortium, 2019d. Identification of novel risk loci, causal insights, and heritable risk for Parkinson’s disease: a meta-analysis of genome-wide association studies. Lancet Neurol. 18, 1091–1102. 10.1016/S1474-4422(19)30320-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, DeStefano AL, Kara E, Bras J, Sharma M, Others, 2014a. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat. Genet. 46, 989–993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nalls MA, Pankratz N, Lill CM, Do CB, Hernandez DG, Saad M, DeStefano AL, Kara E, Bras J, Sharma M, Schulte C, Keller MF, Arepalli S, Letson C, Edsall C, Stefansson H, Liu X, Pliner H, Lee JH, Cheng R, International Parkinson’s Disease Genomics Consortium (IPDGC), Parkinson’s Study Group (PSG) Parkinson's Research: The Organized GENetics Initiative (PROGENI), 23andMe, GenePD, NeuroGenetics Research Consortium (NGRC), Hussman Institute of Human Genomics (HIHG), Ashkenazi Jewish Dataset Investigator, Cohorts for Health and Aging Research in Genetic Epidemiology (CHARGE), North American Brain Expression Consortium (NABEC), United Kingdom Brain Expression Consortium (UKBEC), Greek Parkinson’s Disease Consortium, Alzheimer Genetic Analysis Group, Ikram MA, Ioannidis JPA, Hadjigeorgiou GM, Bis JC, Martinez M, Perlmutter JS, Goate A, Marder K, Fiske B, Sutherland M, Xiromerisiou G, Myers RH, Clark LN, Stefansson K, Hardy JA, Heutink P, Chen H, Wood NW, Houlden H, Payami H, Brice, Scott WK, Gasser T, Bertram L, Eriksson N, Foroud T, Singleton AB, 2014b. Large-scale meta-analysis of genome-wide association data identifies six new risk loci for Parkinson’s disease. Nat. Genet 46, 989–993. 10.1038/ng.3043 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nasser J, Bergman DT, Fulco CP, Guckelberger P, Doughty BR, Patwardhan TA, Jones TR, Nguyen TH, Ulirsch JC, Lekschas F, Mualim K, Natri HM, Weeks EM, Munson G, Kane M, Kang HY, Cui A, Ray JP, Eisenhaure TM, Collins RL, Dey K, Pfister H, Price AL, Epstein CB, Kundaje A, Xavier RJ, Daly MJ, Huang H, Finucane HK, Hacohen N, Lander ES, Engreitz JM, 2021. Genome-wide enhancer maps link risk variants to disease genes. Nature 593, 238–243. 10.1038/s41586-021-03446-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Navarro E, Udine E, de Paiva Lopes K, Parks M, Riboldi G, Schilder BM, Humphrey J, Snijders GJL, Vialle RA, Zhuang M, Sikder T, Argyrou C, Allan A, Chao M, Farrell K, Henderson B, Simon S, Raymond D, Elango S, Ortega RA, Shanker V, Swan M, Zhu CW, Ramdhani R, Walker RH, Tse W, Sano M, Pereira AC, Ahfeldt T, Goate AM, Bressman S, Crary JF, de Witte L, Frucht S, Saunders-Pullman R, Raj T, 2020. Discordant transcriptional signatures of mitochondrial genes in Parkinson’s disease human myeloid cells. bioRxiv. 10.1101/2020.07.20.212407 [DOI] [Google Scholar]
- Nica AC, Montgomery SB, Dimas AS, Stranger BE, Beazley C, Barroso I, Dermitzakis ET, 2010. Candidate causal regulatory effects by integration of expression QTLs with complex trait genetic associations. PLoS Genet. 6, e1000895. 10.1371/journal.pgen.1000895 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nott A, Holtman IR, Coufal NG, Schlachetzki JCM, Yu M, Hu R, Han CZ, Pena M, Xiao J, Wu Y, Keulen Z, Pasillas MP, O’Connor C, Nickl CK, Schafer ST, Shen Z, Rissman RA, Brewer JB, Gosselin D, Gonda DD, Levy ML, Rosenfeld MG, McVicker G, Gage FH, Ren B, Glass CK, 2019. Brain cell type–specific enhancer–promoter interactome maps and disease-risk association. Science 366, 1134–1139. 10.1126/science.aay0793 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noyce AJ, Bandres-Ciga S, Kim J, Heilbron K, Kia D, Hemani G, Xue A, Lawlor DA, Smith GD, Duran R, Gan-Or Z, Blauwendraat C, Gibbs JR, 23andMe Research Team5, International Parkinson’s Disease Genomics Consortium (IPDGC), Hinds DA, Yang J, Visscher P, Cuzick J, Morris H, Hardy J, Wood NW, Nalls MA, Singleton AB, 2019. The Parkinson’s Disease Mendelian Randomization Research Portal. Mov. Disord 34, 1864–1872. 10.1002/mds.27873 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ohnmacht J, May P, Sinkkonen L, Krüger R, 2020. Missing heritability in Parkinson’s disease: the emerging role of non-coding genetic variation. Journal of Neural Transmission. 10.1007/s00702-020-02184-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Padmanabhan S, Polinski NK, Menalled LB, Baptista MAS, Fiske BK, 2019. The Michael J. Fox Foundation for Parkinson’s Research Strategy to Advance Therapeutic Development of PINK1 and Parkin. Biomolecules 9. 10.3390/biom9080296 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Paisán-Ruíz C, Jain S, Evans EW, Gilks WP, Simón J, van der Brug M, López de Munain A, Aparicio S, Gil AM, Khan N, Johnson J, Martinez JR, Nicholl D, Martí Carrera I, Pena AS, de Silva R, Lees A, Martí-Massó JF, Pérez-Tur J, Wood NW, Singleton AB, 2004. Cloning of the gene containing mutations that cause PARK8-linked Parkinson’s disease. Neuron 44, 595–600. 10.1016/j.neuron.2004.10.023 [DOI] [PubMed] [Google Scholar]
- Pande R, Huang Y, Teeple E, Joshi P, Flores-Morales A, Mahieu ML, Pablo Sardi S, Cedazo-Minguez A, Klinger KW, Madden SL, Rajpal D, Kumar D, 2021. Single Cell Atlas of Human Putamen Reveals Disease Specific Changes in Synucleinopathies: Parkinson’s Disease and Multiple System Atrophy. bioRxiv. 10.1101/2021.05.06.442950 [DOI] [Google Scholar]
- Parkinson J, 1817. An essay on the shaking palsy. Whittingham and Rowland, London. [Google Scholar]
- Parkinson’s Disease Genomics, I., 2017. A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nature. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Park JT, Lee Y-S, Cho KA, Park SC, 2018. Adjustment of the lysosomal-mitochondrial axis for control of cellular senescence. Ageing Res. Rev 47, 176–182. 10.1016/j.arr.2018.08.003 [DOI] [PubMed] [Google Scholar]
- Park PJ, 2009. ChIP–seq: advantages and challenges of a maturing technology. Nat. Rev. Genet 10, 669–680. 10.1038/nrg2641 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pasaniuc B, Price AL, 2017. Dissecting the genetics of complex traits using summary association statistics. Nat. Rev. Genet 18, 117–127. 10.1038/nrg.2016.142 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pérez-Palma E, Gramm M, Nürnberg P, May P, Lal D, 2019. Simple ClinVar: an interactive web server to explore and retrieve gene and disease variants aggregated in ClinVar database. Nucleic Acids Res. 47, W99–W105. 10.1093/nar/gkz411 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pers TH, Karjalainen JM, Chan Y, Westra H-J, Wood AR, Yang J, Lui JC, Vedantam S, Gustafsson S, Esko T, Frayling T, Speliotes EK, Genetic Investigation of ANthropometric Traits (GIANT) Consortium, Boehnke M, Raychaudhuri S, Fehrmann RSN, Hirschhorn JN, Franke L, 2015. Biological interpretation of genome-wide association studies using predicted gene functions. Nat. Commun 6, 5890. 10.1038/ncomms6890 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Phani S, Loike JD, Przedborski S, 2012. Neurodegeneration and inflammation in Parkinson’s disease. Parkinsonism Relat. Disord 18 Suppl 1, S207–9. 10.1016/S1353-8020(11)70064-5 [DOI] [PubMed] [Google Scholar]
- Pickrell AM, Youle RJ, 2015. The Roles of PINK1, Parkin, and Mitochondrial Fidelity in Parkinson’s Disease. Neuron. 10.1016/j.neuron.2014.12.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pierce S, Coetzee GA, 2017a. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLOS ONE. 10.1371/journal.pone.0175882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pierce S, Coetzee GA, 2017b. Parkinson’s disease-associated genetic variation is linked to quantitative expression of inflammatory genes. PLoS One 12, e0175882. 10.1371/journal.pone.0175882 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pintacuda G, Lassen FH, Hsu Y-HH, Kim A, Martín JM, Malolepsza E, Lim JK, Fornelos N, Eggan KC, Lage K, 2021. Genoppi is an open-source software for robust and standardized integration of proteomic and genetic data. Nat. Commun 12, 2580. 10.1038/s41467-021-22648-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Poewe W, Seppi K, Tanner CM, Halliday GM, Brundin P, Volkmann J, Schrag A-E, Lang AE, 2017. Parkinson disease. Nat Rev Dis Primers 3, 17013. 10.1038/nrdp.2017.13 [DOI] [PubMed] [Google Scholar]
- Potashkin JA, Blume SR, Runkle NK, 2010. Limitations of animal models of Parkinson’s disease. Parkinsons Dis. 2011, 658083. 10.4061/2011/658083 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Potting C, Crochemore C, Moretti F, Nigsch F, Schmidt I, Manneville C, Carbone W, Knehr J, DeJesus R, Lindeman A, Maher R, Russ C, McAllister G, Reece-Hoyes JS, Hoffman GR, Roma G, Müller M, Sailer AW, Helliwell SB, 2018. Genome-wide CRISPR screen for PARKIN regulators reveals transcriptional repression as a determinant of mitophagy. Proc. Natl. Acad. Sci. U. S. A 115, E180–E189. 10.1073/pnas.1711023115 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pritchard JK, Przeworski M, 2001. Linkage disequilibrium in humans: models and data. Am. J. Hum. Genet 69, 1–14. 10.1086/321275 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raj T, Li YI, Wong G, Humphrey J, Wang M, Ramdhani S, Wang Y-C, Ng B, Gupta I, Haroutunian V, Schadt EE, Young-Pearse T, Mostafavi S, Zhang B, Sklar P, Bennett DA, De Jager PL, 2018. Integrative transcriptome analyses of the aging brain implicate altered splicing in Alzheimer’s disease susceptibility. Nat. Genet 50, 1584–1592. 10.1038/s41588-018-0238-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raj T, Rothamel K, Mostafavi S, Ye C, Lee MN, Replogle JM, Feng T, Lee M, Asinovski N, Frohlich I, Imboywa S, Von Korff A, Okada Y, Patsopoulos NA, Davis S, McCabe C, Paik H-I, Srivastava GP, Raychaudhuri S, Hafler DA, Koller D, Regev A, Hacohen N, Mathis D, Benoist C, Stranger BE, De Jager PL, 2014a. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science 344, 519–523. 10.1126/science.1249547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Raj T, Rothamel K, Mostafavi S, Ye C, Lee MN, Replogle JM, Feng T, Lee M, Asinovski N, Frohlich I, Imboywa S, Von Korff A, Okada Y, Patsopoulos NA, Davis S, McCabe C, Paik H-I, Srivastava GP, Raychaudhuri S, Hafler DA, Koller D, Regev A, Hacohen N, Mathis D, Benoist C, Stranger BE, De Jager PL, 2014b. Polarization of the effects of autoimmune and neurodegenerative risk alleles in leukocytes. Science 344, 519–523. 10.1126/science.1249547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ramdhani S, Navarro E, Udine E, Efthymiou AG, Schilder BM, Parks M, Goate A, Raj T, 2020. Tensor decomposition of stimulated monocyte and macrophage gene expression profiles identifies neurodegenerative disease-specific trans-eQTLs. PLoS Genet. 16, e1008549. 10.1371/journal.pgen.1008549 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Redenšek S, Dolžan V, Kunej T, 2018. From Genomics to Omics Landscapes of Parkinson’s Disease: Revealing the Molecular Mechanisms. OMICS 22, 1–16. 10.1089/omi.2017.0181 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Regev A, Teichmann SA, Lander ES, Amit I, Benoist C, Birney E, Bodenmiller B, Campbell P, Carninci P, Clatworthy M, Clevers H, Deplancke B, Dunham I, Eberwine J, Eils R, Enard W, Farmer A, Fugger L, Göttgens B, Hacohen N, Haniffa M, Hemberg M, Kim S, Klenerman P, Kriegstein A, Lein E, Linnarsson S, Lundberg E, Lundeberg J, Majumder P, Marioni JC, Merad M, Mhlanga M, Nawijn M, Netea M, Nolan G, Pe’er D, Phillipakis A, Ponting CP, Quake S, Reik W, Rozenblatt-Rosen O, Sanes J, Satija R, Schumacher TN, Shalek A, Shapiro E, Sharma P, Shin JW, Stegle O, Stratton M, Stubbington MJT, Theis FJ, Uhlen M, van Oudenaarden A, Wagner A, Watt F, Weissman J, Wold B, Xavier R, Yosef N, Human Cell Atlas Meeting Participants, 2017. The Human Cell Atlas. Elife 6. 10.7554/eLife.27041 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rentzos M, Nikolaou C, Andreadou E, Paraskevas GP, Rombos A, Zoga M, Tsoutsou A, Boufidou F, Kapaki E, Vassilopoulos D, 2007. Circulating interleukin-15 and RANTES chemokine in Parkinson’s disease. Acta Neurol. Scand. 116, 374–379. [DOI] [PubMed] [Google Scholar]
- Reynolds RH, International Parkinson’s Disease Genomics Consortium (IPDGC), Botía J, Nalls MA, Hardy J, Gagliano Taliun SA, Ryten M, System Genomics of Parkinson’s Disease (SGPD), 2019. Moving beyond neurons: the role of cell type-specific gene regulation in Parkinson’s disease heritability. npj Parkinson’s Disease. 10.1038/s41531-019-0076-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Riley EAU, Schekman R, 2021. Point of View: Open science takes on Parkinson’s disease. Elife 10, e66546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robak LA, Jansen IE, van Rooij J, Uitterlinden AG, Kraaij R, Jankovic J, International Parkinson’s Disease Genomics Consortium (IPDGC), Heutink P, Shulman JM, 2017. Excessive burden of lysosomal storage disorder gene variants in Parkinson’s disease. Brain 140, 3191–3203. 10.1093/brain/awx285 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rosenthal LS, Drake D, Alcalay RN, Babcock D, Bowman FD, Chen-Plotkin A, Dawson TM, Dewey RB Jr, German DC, Huang X, Landin B, McAuliffe M, Petyuk VA, Scherzer CR, Hillaire-Clarke CS, Sieber B-A, Sutherland M, Tarn C, West A, Vaillancourt D, Zhang J, Gwinn K, PDBP consortium, 2016. The NINDS Parkinson’s disease biomarkers program. Mov. Disord 31, 915–923. 10.1002/mds.26438 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rotival M, Zeller T, Wild PS, Maouche S, Szymczak S, Schillert A, Castagné R, Deiseroth A, Proust C, Brocheton J, Godefroy T, Perret C, Germain M, Eleftheriadis M, Sinning CR, Schnabel RB, Lubos E, Lackner KJ, Rossmann H, Münzel T, Rendon A, Cardiogenics Consortium, Erdmann J, Deloukas P, Hengstenberg C, Diemert P, Montalescot G, Ouwehand WH, Samani NJ, Schunkert H, Tregouet D-A, Ziegler A, Goodall AH, Cambien F, Tiret L, Blankenberg S, 2011. Integrating genome-wide genetic variations and monocyte expression data reveals trans-regulated gene modules in humans. PLoS Genet. 7, e1002367. 10.1371/journal.pgen.1002367 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ryan SD, Dolatabadi N, Chan SF, Zhang X, Akhtar MW, Parker J, Soldner F, Sunico CR, Nagar S, Talantova M, Lee B, Lopez K, Nutter A, Shan B, Molokanova E, Zhang Y, Han X, Nakamura T, Masliah E, Yates JR, Nakanishi N, Andreyev AY, Okamoto S-I, Jaenisch R, Ambasudhan R, Lipton SA, 2013. Isogenic Human iPSC Parkinson’s Model Shows Nitrosative Stress-Induced Dysfunction in MEF2-PGC1α Transcription. Cell. 10.1016/j.cell.2013.12.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sánchez-Danés A, Richaud-Patin Y, Carballo-Carbajal I, Jiménez-Delgado S, Caig C, Mora S, Di Guglielmo C, Ezquerra M, Patel B, Giralt A, Canals JM, Memo M, Alberch J, López-Barneo J, Vila M, Cuervo AM, Tolosa E, Consiglio A, Raya A, 2012. Disease-specific phenotypes in dopamine neurons from human iPS-based models of genetic and sporadic Parkinson’s disease: iPS cell-based model of Parkinson's disease. EMBO Mol. Med 4, 380–395. 10.1002/emmm.201200215 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schaid DJ, Chen W, Larson NB, 2018. From genome-wide associations to candidate causal variants by statistical fine-mapping. Nat. Rev. Genet 19, 491–504. 10.1038/s41576-018-0016-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schapira AH, Cooper JM, Dexter D, Clark JB, Jenner P, Marsden CD, 1990. Mitochondrial complex I deficiency in Parkinson’s disease. J. Neurochem 54, 823–827. 10.1111/j.1471-4159.1990.tb02325.x [DOI] [PubMed] [Google Scholar]
- Schekman R, Riley EA, 2019. Coordinating a new approach to basic research into Parkinson’s disease. Elife 8. 10.7554/eLife.51167 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schilder BM, Humphrey J, Raj T, 2020. echolocatoR: Automated end-to-end statistical and functional genomic fine-mapping pipeline. bioRxiv. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schilder BM, Raj T, 2020. Fine-mapping of Parkinson’s disease susceptibility loci identifies putative causal variants. bioRxiv. 10.1101/2020.10.22.340158 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schlachetzki JCM, Prots I, Tao J, Chun HB, Saijo K, Gosselin D, Winner B, Glass CK, Winkler J, 2018. A monocyte gene expression signature in the early clinical course of Parkinson’s disease. Sci. Rep 8, 10757. 10.1038/s41598-018-28986-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schulze M, Sommer A, Plötz S, Farrell M, Winner B, Grosch J, Winkler J, Riemenschneider MJ, 2018. Sporadic Parkinson’s disease derived neuronal cells show disease-specific mRNA and small RNA signatures with abundant deregulation of piRNAs. Acta Neuropathologica Communications. 10.1186/s40478-018-0561-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sey NYA, Hu B, Mah W, Fauni H, McAfee JC, Rajarajan P, Brennand KJ, Akbarian S, Won H, 2020. A computational tool (H-MAGMA) for improved prediction of brain-disorder risk genes by incorporating brain chromatin interaction profiles. Nat. Neurosci 23, 583–593. 10.1038/s41593-020-0603-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Simon-Sanchez J, Schulte C, Bras JM, Sharma M, Gibbs JR, Berg D, Paisan-Ruiz C, Lichtner P, Scholz SW, Hernandez DG, Others, 2009. Genome-wide association study reveals genetic risk underlying Parkinson’s disease. Nat. Genet 41, 1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Skene NG, Grant SGN, 2016. Identification of Vulnerable Cell Types in Major Brain Disorders Using Single Cell Transcriptomes and Expression Weighted Cell Type Enrichment. Front. Neurosci 10, 16. 10.3389/fnins.2016.00016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smith AM, Depp C, Ryan BJ, Johnston GI, Alegre-Abarrategui J, Evetts S, Rolinski M, Baig F, Ruffmann C, Simon AK, Hu MTM, Wade-Martins R, 2018. Mitochondrial dysfunction and increased glycolysis in prodromal and early Parkinson’s blood cells. Mov. Disord 33, 1580–1590. 10.1002/mds.104 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smits LM, Reinhardt L, Reinhardt P, Glatza M, Monzel AS, Stanslowsky N, Rosato-Siri MD, Zanon A, Antony PM, Bellmann J, Nicklas SM, Hemmer K, Qing X, Berger E, Kalmbach N, Ehrlich M, Bolognin S, Hicks AA, Wegner F, Sterneckert JL, Schwamborn JC, 2019. Modeling Parkinson’s disease in midbrain-like organoids. NPJ Parkinsons Dis 5, 5. 10.1038/s41531-019-0078-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldner F, Hockemeyer D, Beard C, Gao Q, Bell GW, Cook EG, Hargus G, Blak A, Cooper O, Mitalipova M, Isacson O, Jaenisch R, 2009. Parkinson’s disease patient-derived induced pluripotent stem cells free of viral reprogramming factors. Cell 136, 964–977. 10.1016/j.cell.2009.02.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldner F, Laganière J, Cheng AW, Hockemeyer D, Gao Q, Alagappan R, Khurana V, Golbe LI, Myers RH, Lindquist S, Zhang L, Guschin D, Fong LK, Vu BJ, Meng X, Urnov FD, Rebar EJ, Gregory PD, Zhang HS, Jaenisch R, 2011. Generation of isogenic pluripotent stem cells differing exclusively at two early onset Parkinson point mutations. Cell 146, 318–331. 10.1016/j.cell.2011.06.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Soldner F, Stelzer Y, Shivalila CS, Abraham BJ, Latourelle JC, Barrasa MI, Goldmann J, Myers RH, Young RA, Jaenisch R, 2016. Parkinson-associated risk variant in distal enhancer of α-synuclein modulates target gene expression. Nature 533, 95–99. 10.1038/nature17939 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stypuła G, Kunert-Radek J, Stepień H, Zylińska K, Pawlikowski M, 1996. Evaluation of interleukins, ACTH, cortisol and prolactin concentrations in the blood of patients with parkinson’s disease. Neuroimmunomodulation 3, 131–134. 10.1159/000097237 [DOI] [PubMed] [Google Scholar]
- Takahashi K, Yamanaka S, 2006. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676. 10.1016/j.cell.2006.07.024 [DOI] [PubMed] [Google Scholar]
- Tanigawa Y, Li J, Justesen JM, Horn H, Aguirre M, DeBoever C, Chang C, Narasimhan B, Lage K, Hastie T, Park CY, Bejerano G, Ingelsson E, Rivas MA, 2019. Components of genetic associations across 2,138 phenotypes in the UK Biobank highlight adipocyte biology. Nat. Commun 10, 4064. 10.1038/s41467-019-11953-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tansey MG, Romero-Ramos M, 2018. Immune system responses in Parkinson’s disease: Early and dynamic. European Journal of Neuroscience. 10.1111/ejn.14290 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Teeple E, Joshi P, Pande RS, Huang Y, Karambe A, Latta-Mahieu M, Sardi P, Cedazo-Minguez A, Klinger KW, Flores-Morales A, Madden SL, Rajpal D, Kumar D, 2021. Single Nuclei Sequencing of Human Putamen Oligodendrocytes Reveals Altered Heterogeneity and Disease-Associated Changes in Parkinson’s Disease and Multiple System Atrophy. bioRxiv. 10.1101/2021.05.06.442967 [DOI] [Google Scholar]
- Teves JMY, Bhargava V, Kirwan KR, Corenblum MJ, Justiniano R, Wondrak GT, Anandhan A, Flores AJ, Schipper DA, Khalpey Z, Sligh JE, Curiel-Lewandrowski C, Sherman SJ, Madhavan L, 2017. Parkinson’s Disease Skin Fibroblasts Display Signature Alterations in Growth, Redox Homeostasis, Mitochondrial Function, and Autophagy. Front. Neurosci 11, 737. 10.3389/fnins.2017.00737 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tewhey R, Kotliar D, Park DS, Liu B, Winnicki S, Reilly SK, Andersen KG, Mikkelsen TS, Lander ES, Schaffner SF, Sabeti PC, 2018. Direct Identification of Hundreds of Expression-Modulating Variants using a Multiplexed Reporter Assay. Cell 172, 1132–1134. 10.1016/j.cell.2018.02.021 [DOI] [PubMed] [Google Scholar]
- The Global Parkinson’s Genetics Program, 2021. GP2 : The Global Parkinson’s Genetics Program. Mov. Disord 36, 842–851. 10.1002/mds.28494 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian R, Abarientos A, Hong J, Hashemi SH, Yan R, Dräger N, Leng K, Nalls MA, Singleton AB, Xu K, Faghri F, Kampmann M, 2021. Genome-wide CRISPRi/a screens in human neurons link lysosomal failure to ferroptosis. Nat. Neurosci 10.1038/s41593-021-00862-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tolosa E, Botta-Orfila T, Morató X, Calatayud C, Ferrer-Lorente R, Martí M-J, Fernández M, Gaig C, Raya Á, Consiglio A, Ezquerra M, Fernández-Santiago R, 2018. MicroRNA alterations in iPSC-derived dopaminergic neurons from Parkinson disease patients. Neurobiol. Aging 69, 283–291. 10.1016/j.neurobiolaging.2018.05.032 [DOI] [PubMed] [Google Scholar]
- Townsley KG, Brennand KJ, Huckins LM, 2020. Massively parallel techniques for cataloguing the regulome of the human brain. Nat. Neurosci 23, 1509–1521. 10.1038/s41593-020-00740-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- van der Wijst MGP, de Vries DH, Groot HE, Trynka G, Hon C-C, Bonder M-J, Stegle O, Nawijn MC, Idaghdour Y, van der Harst P, Others, 2020. Science Forum: The single-cell eQTLGen consortium. Elife 9, e52155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wainberg M, Sinnott-Armstrong N, Mancuso N, Barbeira AN, Knowles DA, Golan D, Ermel R, Ruusalepp A, Quertermous T, Hao K, Björkegren JLM, Im HK, Pasaniuc B, Rivas MA, Kundaje A, 2019. Opportunities and challenges for transcriptome-wide association studies. Nat. Genet 51, 592–599. 10.1038/s41588-019-0385-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wang Q, Zhang Y, Wang M, Song W-M, Shen Q, McKenzie A, Choi I, Zhou X, Pan P-Y, Yue Z, Zhang B, 2019. The landscape of multiscale transcriptomic networks and key regulators in Parkinson’s disease. Nature Communications. 10.1038/s41467-019-13144-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- Watanabe K, Taskesen E, van Bochoven A, Posthuma D, 2017. Functional mapping and annotation of genetic associations with FUMA. Nat. Commun 8, 1826. 10.1038/s41467-017-01261-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weissbrod O, Hormozdiari F, Benner C, Cui R, Ulirsch J, Gazal S, Schoech AP, van de Geijn B, Reshef Y, Márquez-Luna C, O’Connor L, Pirinen M, Finucane HK, Price AL, 2020. Functionally informed fine-mapping and polygenic localization of complex trait heritability. Nat. Genet 52, 1355–1363. 10.1038/s41588-020-00735-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams A, 1984. MPTP parkinsonism. Br. Med. J 289, 1401–1402. 10.1136/bmj.289.6456.1401 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wirdefeldt K, Gatz M, Reynolds CA, Prescott CA, Pedersen NL, 2011. Heritability of Parkinson disease in Swedish twins: a longitudinal study. Neurobiol. Aging 32, 1923.e1–8. 10.1016/j.neurobiolaging.2011.02.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Witoelar A, Jansen IE, Wang Y, Desikan RS, Gibbs JR, Blauwendraat C, Thompson WK, Hernandez DG, Djurovic S, Schork AJ, Bettella F, Ellinghaus D, Franke A, Lie BA, McEvoy LK, Karlsen TH, Lesage S, Morris HR, Brice A, Wood NW, Heutink P, Hardy J, Singleton AB, Dale AM, Gasser T, Andreassen OA, Sharma M, International Parkinson’s Disease Genomics Consortium (IPDGC), North American Brain Expression Consortium (NABEC), and United Kingdom Brain Expression Consortium (UKBEC) Investigators, 2017. Genome-wide Pleiotropy Between Parkinson Disease and Autoimmune Diseases. JAMA Neurol. 74, 780–792. 10.1001/jamaneurol.2017.0469 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wong AK, Krishnan A, Troyanskaya OG, 2018. GIANT 2.0: genome-scale integrated analysis of gene networks in tissues. Nucleic Acids Res. 46, W65–W70. 10.1093/nar/gky408 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang H, Liu D, Zhao C, Feng B, Lu W, Yang X, Xu M, Zhou W, Jing H, Yang J, 2021. Mendelian randomization integrating GWAS and eQTL data revealed genes pleiotropically associated with major depressive disorder. Transl. Psychiatry 11, 225. 10.1038/s41398-021-01348-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Young AMH, Kumasaka N, Calvert F, Hammond TR, Knights A, Panousis N, Schwartzentruber J, Liu J, Kundu K, Segel M, Murphy N, McMurran CE, Bulstrode H, Correia J, Budohoski KP, Joannides A, Guilfoyle MR, Trivedi R, Kirollos R, Morris R, Garnett MR, Fernandes H, Timofeev I, Jalloh I, Holland K, Mannion R, Mair R, Watts C, Price SJ, Kirkpatrick PJ, Santarius T, Soranzo N, Stevens B, Hutchinson PJ, Franklin RJM, Gaffney DJ, 2021. A map of transcriptional heterogeneity and regulatory variation in human microglia. Nat. Genet 2019.12.20.874099. 10.1038/s41588-021-00875-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou J, Theesfeld CL, Yao K, Chen KM, Wong AK, Troyanskaya OG, 2018. Deep learning sequence-based ab initio prediction of variant effects on expression and disease risk. Nat. Genet. 50, 1171–1179. 10.1038/s41588-018-0160-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou J, Troyanskaya OG, 2015. Predicting effects of noncoding variants with deep learning–based sequence model. Nat. Methods 12, 931–934. 10.1038/nmeth.3547 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimprich A, Biskup S, Leitner P, Lichtner P, Farrer M, Lincoln S, Kachergus J, Hulihan M, Uitti RJ, Calne DB, Stoessl AJ, Pfeiffer RF, Patenge N, Carbajal IC, Vieregge P, Asmus F, Müller-Myhsok B, Dickson DW, Meitinger T, Strom TM, Wszolek ZK, Gasser T, 2004. Mutations in LRRK2 cause autosomal-dominant parkinsonism with pleomorphic pathology. Neuron 44, 601–607. 10.1016/j.neuron.2004.11.005 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.