Complexities of Understanding Function from CKD-Associated DNA Variants : Clinical Journal of the American Society of Nephrology

Journal Logo

Genomics of Kidney Disease

Complexities of Understanding Function from CKD-Associated DNA Variants

Lin, Jennie1,2; Susztak, Katalin3,4

Author Information
CJASN 15(7):p 1028-1040, July 2020. | DOI: 10.2215/CJN.15771219
  • Open

Abstract

Overview of Genome-Wide Association Studies

CKD, and by extension kidney function, has a strong genetic basis, with heritability of eGFR and albuminuria calculated to be around 16%–50% (1,2). Since the early 2000s, genome-wide association studies (GWASs) have been a powerful platform for identifying associations between common disease development and common DNA variants in the human population. These studies have identified hundreds of genetic regions, or loci, where nucleotide sequences differ between those with disease and those without disease (3–4567891011). Because these studies survey the entire human genome, many of their reported genetic variants lie within loci not previously studied in the context of the disease of interest. For example, the first GWAS for CKD in 2009 and a larger GWAS for CKD in 2016 identified dozens of novel loci for CKD (4,12). Most (close to 90%) of the genetic variants associated with development of complex disease are located in noncoding regions of the genome (13,14), and they are distinct from genetic variants associated with Mendelian, or monogenic, diseases. Each DNA variant associated with complex CKD or kidney-relevant traits exerts a small effect on disease pathogenesis. As such, small effects from numerous DNA variants in aggregate drive the incidence of complex CKD and are best detected in a large sample size in population studies such as the latest GWAS for CKD, which involved over 1 million individuals and detected more than 300 loci for kidney function (10,15).

Although most GWAS data are not yet ready for direct development of novel therapeutics, they must highlight critical disease-causing genes for experimental studies and eventual downstream therapeutic targeting. This concept forms the basis of functional genomics, which leverages unbiased genetic data to assign function to DNA variants that are causal in disease mechanisms (Figure 1). Ideally, the GWAS functional genomics workflow would result in the development of novel therapeutic strategies for complex diseases such as CKD. However, despite the many genetic regions reported to associate with disease traits, only a handful of GWAS loci and variants have been shown to be causally linked to disease development, emphasizing the importance of interrogating these loci for function. In this review, we introduce concepts relevant to understanding GWAS, bioinformatics approaches for prioritizing GWAS variants for functional validation, and experimental approaches for validating causality.

fig1
Figure 1.:
Genome-wide association analysis. Human genome-wide association studies (GWASs) compare single nucleotide polymorphism frequency in healthy control and disease patients (A). Genetic variation (B) in many loci show association with kidney disease development, as seen in a GWAS Manhattan plot (C). Within a GWAS locus, many variants show significant association with disease development, as seen in a locus zoom plot (D). The causal variant, the target gene, the target cell type, and the disease mechanism understanding are critical to translate GWAS discoveries into clinically actionable functions.

Basic Study Design

For a glossary of the terms used in this overview, see Table 1. The purpose of a GWAS is to determine which genomic loci are associated with a particular complex disease or trait. Typically, the variants tested are single nucleotide polymorphisms (SNPs), millions of which can be assayed at the same time using SNP arrays, and more recently, whole-genome sequencing (16). DNA variants can also include insertions and deletions, although these are less common. If the queried phenotype is dichotomous (present or absent), a GWAS will evaluate whether an SNP’s allele frequency differs between a group of cases and a group of controls (Figure 1). If the disease phenotype is a continuous quantitative trait such as eGFR, a GWAS will evaluate whether there are statistically significant differences in the trait among the groups of people with the three different genotypes at the SNP. These genotypes include being homozygous for the major (or more common) allele, homozygous for the minor allele, or heterozygous for both (17). Robust statistical significance for a GWAS is typically P<5×10−8 because of a stringent Bonferroni correction for multiple testing accounting for the roughly 1 million independent SNPs tested simultaneously. The corrected P value is obtained by dividing the designated α (0.05) by the number of tests performed (1 million, because of testing 1 million independent SNPs): 0.05/106=5×10−8.

Table 1. - Glossary of terms
Term Definition
Allele A variant form of a gene’s locus, often refers to alternative nucleotide at an SNP location
Bonferroni correction A type of statistical correction for multiple comparisons
Causal Contributing to causing a disease or trait
Chromatin Substance within a chromosome consisting of DNA and protein
Enhancer Short genomic DNA sequence where transcriptional modifiers can bind
Linkage disequilibrium (LD) Nonrandom association of one variant with another
Locus Genomic region flanked by recombination hotspots
Plasmid Circular DNA. In research, it is introduced into cells for transgenic expression of the cargo sequence
Single nucleotide polymorphism (SNP) A sequence variant consisting of a difference in a single nucleotide
Transcription factor A protein that controls rate of transcription by binding to DNA sequences

Linkage Disequilibrium and Minor Allele Frequency

During meiosis, relatively large regions of the human genome are inherited together. Genes and DNA variants located within these regions are therefore “linked”. Because of genetic linkage, two SNPs (that are usually close to each other) can have a high degree of linkage disequilibrium (LD), which is the nonrandom association of alleles (Figure 1). For an SNP to be in high LD with another SNP, the minor allele of one SNP is often inherited with the minor allele of the other SNP (17). Close correlation among DNA variants, owing to genetic linkage and LD, has been essential for the success of GWAS efforts, which leverage the limited number of independent LDs to calculate the association between an SNP and a disease or trait. Although the term GWAS includes the words “genome-wide,” not all SNPs in the human genome are captured by commercial genotyping microarray platforms typically used in a GWAS. As multiple SNPs are inherited together, it is sufficient to genotype one or a few SNPs from each independent LD, whereas the rest of the variants can be estimated on the basis of their LD. The tag-SNPs on these microarray platforms are chosen for genotyping purposes because they have a large amount of LD with neighboring SNPs (18).

Another important issue to consider is the minor allele frequency (MAF). As a GWAS looks for a statistical difference of genetic variation between cases and control, statistical power to detect significant associations between variant and trait is partially a function of MAF. A lower MAF for an allele (e.g., a rarer variant) reduces the number of individuals available for association analysis and thus makes detection of an association less likely unless that variant exerts a large phenotypic effect. Of note, the MAF of an SNP is calculated by population and thus may vary among different ethnicities with contrasting evolutionary histories (9,17). These ancestry-specific MAFs correlate with differences in disease prevalence among populations. For example, IgA nephropathy is more commonly seen in individuals of East Asian and European descent, and risk variants associated with IgA nephropathy are higher in frequency in these populations (19). Another example of how ancestry-related differences in MAFs can drive population-based differences in disease prevalence is apo L1 (APOL1) nephropathy. The G1 and G2 disease risk alleles for nondiabetic CKD are common among individuals of African ancestry but rare in other ethnicities; they are also inherited in an ancestry-specific haplotype (20,21). These risk alleles have been validated to drive a portion of the excess CKD risk seen in the black population (20,22–2324). It is therefore very important to adjust the genetic data for population ancestry before conducting genotype–disease association because ancestry-driven allele frequency changes can bias the results (25), highlighting the importance of cohort demographics when designing, conducting, and interpreting a GWAS.

From Association to Function

A GWAS reports the associations between variants and disease trait. Because genetic associations do not prove causality, “post-GWAS” work must be performed to interrogate these associations for function and identify the variants and genes that are causal, or directly and biologically relevant, for the disease of interest (Figure 1). In a GWAS, an association between a tag-SNP and an inherited trait or disease may be indirect if the causal variant is in high LD with the tag-SNP. Within a locus, additional fine mapping, the methods for which are beyond the scope of this review (26), may identify select candidate causal SNPs for functional validation. As mentioned above, only a handful of GWAS-identified loci have been causally linked to disease. In these regions, the causal variant is usually located within a regulatory region specific to cell type. These regions typically include enhancers, which are short sequences of DNA that interact with transcription factors to increase the likelihood that a gene is transcribed into mRNA. A causal nucleotide change in an enhancer region alters transcription factor binding strength, resulting in a small but measurable change in mRNA transcript and protein levels of a causal gene that contributes to disease development. Correct identification of causal variants and causal genes then allows for opportunities to interrogate the mechanisms by which those genes may alter the phenotype of interest.

Post-GWAS Validation of Causal Variants and Causal Genes

A major GWAS challenge stems from having many DNA variants inherited together with some degree of LD. Because hundreds or even thousands of SNPs could be present at a specific genetic locus, identifying causal variants and causal genes is extremely difficult and has only been reported to less than a handful of loci. In addition careful bioinformatics analyses to narrow down candidate variants, cellular and in vivo studies are needed to demonstrate that these variants and their target genes are directly causal, or relevant to disease pathogenesis. Here, we highlight some commonly used approaches to prioritize, identify, and functionally validate causal variants and causal genes in disease development (Figure 1).

Causal Variants

As mentioned above, many DNA variants are inherited together with varying degrees of LD. One approach to narrowing down the genomic “neighborhood” of the causal variant(s) is to leverage epigenetic and epigenomic data, which help mark functionally important areas in the genome. Functionally active areas in the genome are usually defined by open chromatin where DNA is not tightly bound to histones and where transcription factors can bind and RNA can be transcribed (Figure 2). Of note, patterns of open chromatin tend to be specific to cell type (18,25), so an area of open chromatin in a proximal tubular cell may not be open in a hepatocyte or even a podocyte. One method to map open chromatin within a cell type is to perform a DNase I hypersensitivity assay, in which DNA fragments are digested out for high-throughput sequencing (27). However, this method is technically difficult, and more investigators are using assay for transposase-accessible chromatin using sequencing, which involves tagging regions of open chromatin with hyperactive Tn5 transposases and sequencing these purified DNA fragments (28). In addition to using open chromatin maps, cell type–specific epigenetic data from chromatin immunoprecipitation followed by sequencing can be leveraged to determine whether candidate SNPs lie within promoter, enhancer, or repressor regions (29,30). For example, histone tail modification marks such as monomethylated H3K4 and acetylated H3K27 indicate active enhancer regions (31). These epigenome maps for various (nonkidney) cell types and tissues are publicly available through the Encyclopedia of Coding Elements and International Human Epigenome Consortium projects (32,33). Comparing GWAS loci with the open chromatin and histone maps can narrow the window of candidate SNPs to prioritize, although this approach cannot fully validate causality of these variants.

fig2
Figure 2.:
Epigenomic annotation of genomic locus to prioritize candidate causal variants. Large amounts of the noncoding genome have regulatory function (A). These regions include the promoter, enhancer, or insulator that regulate the transcribed region. Regulatory regions can be identified by epigenome mapping as specific histone tail modifications are enriched on specific regulatory elements. Candidate causal variants (purple stars) often fall within these regions (B). DNA folds such that the enhancer region (yellow) interacts with the promoter region (red) to increase transcription of a gene. These spatial regulatory relationships can be captured through molecular biology techniques (C). Although show association with kidney disease development (top graph of single nucleotide polymorphisms [SNPs] above dashed line indicating statistical significance), only a portion of this region demonstrates regulatory activity in kidney tubule cells. The bottom plots are visualizations of enhancer-specific histone tail modification from chromatin immunoprecipitation sequencing of human kidney and macrophages, revealing cell and tissue specificity of transcriptional regulation.

To drill down further on whether candidate variants are causal in regulating gene expression, experimental reporter assays can be performed. A reporter assay consists of inserting a DNA sequence containing the candidate SNP either upstream or downstream of a promoter reporter gene cassette in a DNA plasmid. The plasmid is introduced into the cell type of interest, which expresses the relevant proteins regulating transcription, such as transcription factors, which often are specific to cell type. If the SNP alters transcriptional efficiency by affecting the binding of transcription factors, its presence will change the expression level of the reporter protein, such as luciferase or green fluorescent protein (17). This type of reporter assay can interrogate a portion of one locus at a time. Increasing throughput and efficiency, the massively parallel reporter assay (MPRA) can be used to assay hundreds or thousands of DNA regions simultaneously (34). The MPRA leverages the use of unique barcodes for each DNA region represented by an oligonucleotide, and these regions with their barcodes are cloned into reporter plasmids. Cells that have alterations in reporter expression are then sorted and sequenced for the barcodes to identify SNPs in regions that regulate transcription. The major limitation of using either a single reporter assay or the MPRA approach is that the transcriptional interrogation occurs not in the native genomic or epigenomic context of the candidate SNPs. The assayed regions are shorter than the full regulatory sequences at the native genomic loci, and in being cloned into plasmids they may also change the native chromatin structure that plays a key role in gene transcription.

To circumvent the challenges and shortcomings of reporter assays, candidate SNPs and loci can be interrogated in their native genomic context through genome editing. Although several methods for genome editing exist, CRISPR-Cas9 has taken center stage as a feasible and accessible method to knock in candidate SNPs at their natural chromosomal position. A detailed primer on CRISPR-Cas9 can be reviewed elsewhere (35,36). Briefly, the CRISPR-Cas9 system leverages a guide RNA that directs a nuclease enzyme to make a cut in the genomic DNA. Because the guide RNA is derived from a carefully designed and engineered oligonucleotide, the nuclease action is very precise in location. The break in genomic DNA is then repaired either (1) through nonhomologous end joining or (2) homology-directed repair. Nonhomologous end joining is an imperfect process by which the cut ends of the genomic DNA are religated without a template sequence to guide repair. Without such a template, additional nucleotides may be added, or existing nucleotides excluded, in the repair process, thereby potentially causing a frame shift that knocks out expression of that gene. Unlike nonhomologous end joining, homology-directed repair involves an exogenous homologous template sequence that can bind to the cut blunt ends. To introduce a variant or SNP into the genome, that homologous template contains the desired sequence, which then gets incorporated into the genomic DNA during the repair process. When a desired SNP is introduced into a kidney cell, for example, differences in expression of nearby genes can be measured compared with an isogenic control, and other cellular phenotypes can also be evaluated.

Causal Genes

To investigate potential novel pathways involved in disease pathogenesis, the genes regulated by causal variants need to be identified (Figure 3). One approach to identify causal genes is through expression quantitative trait locus (eQTL) mapping, which assesses whether a candidate SNP modulates expression of nearby genes (cis regulation) or distant genes on the same or different chromosome (trans regulation). An eQTL is a gene–SNP pair for which a gene’s expression associates with the allelic configuration of the SNP. As such, eQTL analysis requires both the genotype and gene expression data for cells of tissues of interest.

fig3
Figure 3.:
Expression of quantitative trait locus (eQTL) mapping to identify causal genes GWAS reports association between genetic variants and disease development. A variant identified in GWAS will then undergo eQTL mapping to identify which gene(s) that variant regulates in terms of transcriptional abundance. eQTL mapping has demonstrated that variant effects on gene expression can be specific to compartments of the nephron. In this figure, the risk allele, B, located in the yellow oval region increases expression of the eGene measured in tubules, as shown in the box plot of gene expression level (y axis) and genotype (x axis). However, the B allele does not alter expression of that same gene in the glomerulus. This is consistent with tubule-specific eQTL.

One of the largest data sets to incorporate both genotype and transcriptomic data are from the Genotype-Tissue Expression (GTEx) project, which characterized gene expression levels across hundreds of postmortem donors and over 40 tissues in the human body (37). GTEx data have facilitated eQTL analyses for a variety of diseases and traits, including schizophrenia, coronary artery disease, and CKD (38–39404142). These analyses have highlighted potential candidate causal variants and causal genes, although the limited number of kidney samples has limited the power of eQTL analyses for CKD. In addition, RNA sequencing (RNA-seq) data for GTEx samples reflect whole tissue that includes different cell types if different organs can have different expression patterns and eQTLs, can different cell types within an organ also have different eQTLs from each other?

With at least 26 different cell types in the kidney (43) and at least 16 detected by single-cell RNA-sequencing (44–4546), interrogating how eQTLs differ among these cell types will help prioritize key candidate causal variants and target genes for functional validation and mechanistic insights. Although eQTL data from single-cell RNA-sequencing of kidneys are not yet available, an eQTL atlas for the glomerular and tubular compartments of the human kidney has revealed the existence of compartment-specific eQTLs not seen in whole kidney data (42,47). Because CKD spans many different diseases—some with glomerular pathophysiology and others with significant tubular and interstitial pathology—eQTLs specific to each compartment reflect the expected differences in disease pathogenesis. In addition, CKD GWAS loci tend to associate with compartment-specific eQTLs and are further away from promoter regions, suggesting that compartment-specific eQTLs may indeed act in enhancer regions (42). The concept of cell-specific or compartment-specific eQTL is not limited to the kidney: studies of other organ systems, such as skin and adipose, also highlight the importance of cell-specific programs of transcriptional regulation (48,49). Because GWAS SNPs and even subthreshold GWAS SNPs (that do not meet genome-wide significance) tend to map to regulatory regions and have been shown to modulate transcriptional activity (50,51), knowledge of cell specificity of these regulatory features can provide clues for causal cell types and pathophysiology. Taken together with high-resolution epigenomic data that are also cell type–specific, evaluation of the causal cell type for a disease-associated variant will assist in validating candidate variants and target genes.

Although expression of quantitative trait loci (QTLs) is currently most widely used to annotate GWAS loci, multiple additional molecular QTLs can be used to annotate genetic regions, such as histone QTLs, accessible chromatin QTLs, methylation QTLs, protein QTLs, or metabolite QTLs (52–5354). The concept around additional molecular QTLs is the same: they are based on the generation of an external database where genetic variations and other molecular trait associations are established. Integration of the GWAS and multiple molecular traits are critical for gene prioritization. Variants that are associated both with disease states and influence molecular traits are usually prioritized for further analysis.

In addition to QTLs, another approach to identifying causal genes, especially for variants in noncoding regions that could be hundreds of kilobases away from their target genes, is to test physical associations between candidate SNPs and promoter regions. Chromosomes can bend and fold in different conformations that allow for distal regulatory elements, such as enhancers housing GWAS SNPs, to make direct contact with promoters, thereby modulating transcription of putative target genes. These looping interactions can be interrogated through experimental methods known as chromosome conformation capture accompanied by high-throughput sequencing, which can range from large-scale genome-wide assays to examining where one enhancer region binds (55,56). Indeed, chromosome capture has been instrumental in our understanding of GWAS SNP–gene interactions for obesity and kidney (57,58).

Once candidate target genes of causal variants are identified, further mechanistic studies to evaluate functional significance of these genes can be performed. Experimental approaches for studying kidney GWAS traits include genetic deletion of candidate genes in zebrafish (4,59) and mice (42) to evaluate whether altering expression of those genes induces or protects against kidney injury. Once a phenotype is established from perturbation of candidate gene expression, mechanistic studies to test how these genes contribute to CKD and other kidney-relevant traits can provide new insights into disease pathogenesis and reveal potential novel therapeutic strategies.

Examples of Functional Follow-Up of Kidney Disease–Associated Variants

Hundreds of GWAS loci have been reported to associate with CKD and other kidney traits. However, determining whether these loci have functional significance in kidney physiology and disease requires probing beyond bioinformatics analyses and performing experimental science. To illustrate how the link between genotype and kidney phenotype has been explored, below we review the functional studies examining noncoding variants from two loci, SHROOM3 and DAB2, that have been associated with kidney traits in multiple GWAS reports (3,4,12,60).

SHROOM3

GWASs of largely European ancestry individuals have linked the SNP rs17319721 in an intronic region of SHROOM3 with CKD and eGFR (4,60), although the role of SHROOM3 expression had not previously been established for CKD. Menon et al. demonstrated through integration of human patient data, reporter assays, and in vivo experiments that the risk allele at the SHROOM3 locus increases SHROOM3 expression through increased binding of a transcription factor TCF7L2 in an Wnt/β-catenin–dependent fashion. They showed that the risk allele increases SHROOM3 expression in kidney allograft samples, and that the SNP overlies a region with a consensus binding sequence for TCF7L2 that then activates SHROOM3 transcription (61). In addition, the risk allele and higher SHROOM3 expression correlated with higher chronic allograft nephropathy fibrosis scores (61). In a complementary experiment, mice undergoing unilateral ureteral obstruction to induce fibrosis had attenuation of profibrotic signaling on knockdown of Shroom3 both in a whole-body knockdown model and a tubule-specific overexpression model (61).

These results differ somewhat from other functional studies. Prokop et al. demonstrated that introducing wild-type Shroom3 into the fawn-hooded hypertensive rat, which develops spontaneous proteinuric disease, improved glomerular sclerosis. This directionality conflicts with the study by Menon et al. In addition, the same group later published that the rs17319721 risk allele does increase expression of a shorter isoform of SHROOM3, but that TCF7L2 acts as a repressor that gets released in the presence of the risk allele (62). These differences in proposed mechanisms are likely because of different model systems, divergent methods for interrogating the risk variant (reporter assays versus genome editing), and different cultured cells such as tubule cells and podocytes. Indeed, establishing a robust and high-fidelity model system is a key component of functional validation of GWAS loci. Differences among model systems, as well as between humans and mice, have led to conflicting study results for other kidney loci, such as UMOD (63,64) and APOL1 (65,66). Although the SHROOM3 studies affirm that the risk allele modulates SHROOM3 expression, they also highlight the importance of methodology in mechanistic interrogation of candidate SNPs and target genes.

DAB2

Further illustrating the importance of cell types in studying variants is the DAB2-C9 locus. GWAS for kidney traits identified an association between rs11959928 and CKD (3,4). However, on whole-kidney eQTL analysis by Ko et al. (59) the SNP did not associate with expression levels of nearby genes DAB2 or C9. Upon closer examination with compartment-specific eQTL analysis, the risk allele for rs11959928 did associate with tubular expression of DAB2 but not C9, and there were no significant eQTLs for this SNP in the glomerular compartment, highlighting the importance of cell specificity in eQTL studies of complex tissues (Figure 4) (42). In addition, although DAB2 is expressed in multiple tissues, the specific genetic variant only influenced the level of DAB2 in kidney tubules and not in other tissues.

fig4
Figure 4.:
Identification of DAB2 as a kidney disease gene. Prior GWAS for kidney disease traits identified an association between the DAB2-C9 locus and eGFR. The tag SNP is located between the two genes, leading to the questions of which gene is regulated by a causal variant or variants, which cell type is relevant to the association, and what the effect of the regulated gene is on kidney disease. Compartment-specific eQTL mapping of the GWAS variant revealed that DAB2 expression is modulated by the variant in the tubule portion of the nephron. C9 expression levels were not influenced by the variant. To confirm DAB2’s role in tubule health, tubule-specific knockout mice for Dab2 and C9 were generated and stressed with folic acid, which induces fibrotic injury. The Dab2 tubule-specific knockout mice developed less fibrosis than the wild-type control mice, suggesting that mice with genetic lowering of DAB2 protein is protected from development of kidney disease. No effect was seen in the C9 knockout mice.

To drill down further on whether DAB2 or C9 is the causal gene, Qiu et al. examined the response to profibrotic injury in tubule-specific knockdown of these genes in mice. No differences were seen in the C9 knockdown mice, but the Dab2 knockdown mice demonstrated partial protection against profibrotic injury states (Figure 4). Primary cell culture experiments showed that less Dab2 expression led to less TGFβ-induced Smad2 and Smad3 phosphorylation and less TGFβ-induced fibronectin and collagen1 production (42). Without the level of resolution compartment-specific or single-cell analyses offer, the causal gene would not have been identified.

Challenges and Moving Forward

In this era of high-throughput sequencing and massive amounts of data collected on the human genome, multiple GWAS efforts have probed the genetic basis of CKD, revealing many novel loci that associate with kidney function and other kidney-related traits. However, only a few of these CKD-relevant loci have yielded published functional studies, some with conflicting results. With the causal variants likely exerting only small effects on kidney function, recapitulating disease phenotype experimentally by interrogating SNPs from one locus may be difficult. The GWAS field is moving toward development of polygenic risk scores for risk prediction of disease (67–6869), aligning with the concept that an aggregate of variants exerting small effects contributes to disease development. Furthermore, CKD, eGFR, and albuminuria are broad traits encompassing heterogeneous disease causes and overlapping presentations. Because the strength of association between genotype and phenotype relies on the quality of phenotype characterization, phenotypes based on serum creatinine, which can vary in accuracy among centers (70), may limit functional validation.

Even with further optimization of genetic epidemiology studies of CKD, assigning function to variants will require integrative and cell-specific approaches, as evident by the DAB2 example presented above. Merging genetic data with epigenetics and transcriptomics helps identify causal variants and causal genes involved in CKD pathogenesis in a cell type–specific manner. To ensure success, these data from human kidney biobanks will be instrumental, and a coordinated effort among CKD investigators will be needed for biospecimen collection and appropriate processing for the technologies relevant to epigenetics and single-cell transcriptomics. With these collaborative efforts in place, further experimental studies with cellular and in vivo models will be needed to validate causal candidates, and are essential to elucidating novel kidney disease mechanisms for eventual therapeutic targeting.

Disclosures

Dr. J. Lin and Dr. K. Susztak report work in the laboratory is supported by Boehringer Ingelheim, Gilead, GSK, Merck, Bayer and Regeneron. Dr. K. Susztak has consulted for Janssen, Chemocentryx, Maze Pharma, and Jnana.

Funding

Dr. J. Lin is supported by National Institutes of Health grant K08 HL135348, and the laboratory of Dr. K. Susztak is supported by National Institutes of Health grants R01 DK076077, DK087635, and DK105821.

Published online ahead of print. Publication date available at www.cjasn.org.association.

References

1. Fox CS, Yang Q, Cupples LA, Guo C-Y, Larson MG, Leip EP, Wilson PWF, Levy D: Genomewide linkage analysis to serum creatinine, GFR, and creatinine clearance in a community-based population: The Framingham Heart Study. J Am Soc Nephrol 15: 2457–2461, 2004 15339995
2. Fox CS, Yang Q, Guo C-Y, Cupples LA, Wilson PWF, Levy D, Meigs JB: Genome-wide linkage analysis to urinary microalbuminuria in a community-based sample: The Framingham Heart Study. Kidney Int 67: 70–74, 2005 15610229
3. Köttgen A, Pattaro C, Böger CA, Fuchsberger C, Olden M, Glazer NL, Parsa A, Gao X, Yang Q, Smith AV, O’Connell JR, Li M, Schmidt H, Tanaka T, Isaacs A, Ketkar S, Hwang S-J, Johnson AD, Dehghan A, Teumer A, Paré G, Atkinson EJ, Zeller T, Lohman K, Cornelis MC, Probst-Hensch NM, Kronenberg F, Tönjes A, Hayward C, Aspelund T, Eiriksdottir G, Launer LJ, Harris TB, Rampersaud E, Mitchell BD, Arking DE, Boerwinkle E, Struchalin M, Cavalieri M, Singleton A, Giallauria F, Metter J, de Boer IH, Haritunians T, Lumley T, Siscovick D, Psaty BM, Zillikens MC, Oostra BA, Feitosa M, Province M, de Andrade M, Turner ST, Schillert A, Ziegler A, Wild PS, Schnabel RB, Wilde S, Munzel TF, Leak TS, Illig T, Klopp N, Meisinger C, Wichmann H-E, Koenig W, Zgaga L, Zemunik T, Kolcic I, Minelli C, Hu FB, Johansson A, Igl W, Zaboli G, Wild SH, Wright AF, Campbell H, Ellinghaus D, Schreiber S, Aulchenko YS, Felix JF, Rivadeneira F, Uitterlinden AG, Hofman A, Imboden M, Nitsch D, Brandstätter A, Kollerits B, Kedenko L, Mägi R, Stumvoll M, Kovacs P, Boban M, Campbell S, Endlich K, Völzke H, Kroemer HK, Nauck M, Völker U, Polasek O, Vitart V, Badola S, Parker AN, Ridker PM, Kardia SLR, Blankenberg S, Liu Y, Curhan GC, Franke A, Rochat T, Paulweber B, Prokopenko I, Wang W, Gudnason V, Shuldiner AR, Coresh J, Schmidt R, Ferrucci L, Shlipak MG, van Duijn CM, Borecki I, Krämer BK, Rudan I, Gyllensten U, Wilson JF, Witteman JC, Pramstaller PP, Rettig R, Hastie N, Chasman DI, Kao WH, Heid IM, Fox CS: New loci associated with kidney function and chronic kidney disease. Nat Genet 42: 376–384, 2010 20383146
4. Pattaro C, Teumer A, Gorski M, Chu AY, Li M, Mijatovic V, Garnaas M, Tin A, Sorice R, Li Y, Taliun D, Olden M, Foster M, Yang Q, Chen M-H, Pers TH, Johnson AD, Ko Y-A, Fuchsberger C, Tayo B, Nalls M, Feitosa MF, Isaacs A, Dehghan A, d’Adamo P, Adeyemo A, Dieffenbach AK, Zonderman AB, Nolte IM, van der Most PJ, Wright AF, Shuldiner AR, Morrison AC, Hofman A, Smith AV, Dreisbach AW, Franke A, Uitterlinden AG, Metspalu A, Tönjes A, Lupo A, Robino A, Johansson Å, Demirkan A, Kollerits B, Freedman BI, Ponte B, Oostra BA, Paulweber B, Krämer BK, Mitchell BD, Buckley BM, Peralta CA, Hayward C, Helmer C, Rotimi CN, Shaffer CM, Müller C, Sala C, van Duijn CM, Saint-Pierre A, Ackermann D, Shriner D, Ruggiero D, Toniolo D, Lu Y, Cusi D, Czamara D, Ellinghaus D, Siscovick DS, Ruderfer D, Gieger C, Grallert H, Rochtchina E, Atkinson EJ, Holliday EG, Boerwinkle E, Salvi E, Bottinger EP, Murgia F, Rivadeneira F, Ernst F, Kronenberg F, Hu FB, Navis GJ, Curhan GC, Ehret GB, Homuth G, Coassin S, Thun G-A, Pistis G, Gambaro G, Malerba G, Montgomery GW, Eiriksdottir G, Jacobs G, Li G, Wichmann H-E, Campbell H, Schmidt H, Wallaschofski H, Völzke H, Brenner H, Kroemer HK, Kramer H, Lin H, Leach IM, Ford I, Guessous I, Rudan I, Prokopenko I, Borecki I, Heid IM, Kolcic I, Persico I, Jukema JW, Wilson JF, Felix JF, Divers J, Lambert J-C, Stafford JM, Gaspoz J-M, Smith JA, Faul JD, Wang JJ, Ding J, Hirschhorn JN, Attia J, Whitfield JB, Chalmers J, Viikari J, Coresh J, Denny JC, Karjalainen J, Fernandes JK, Endlich K, Butterbach K, Keene KL, Lohman K, Portas L, Launer LJ, Lyytikäinen L-P, Yengo L, Franke L, Ferrucci L, Rose LM, Kedenko L, Rao M, Struchalin M, Kleber ME, Cavalieri M, Haun M, Cornelis MC, Ciullo M, Pirastu M, de Andrade M, McEvoy MA, Woodward M, Adam M, Cocca M, Nauck M, Imboden M, Waldenberger M, Pruijm M, Metzger M, Stumvoll M, Evans MK, Sale MM, Kähönen M, Boban M, Bochud M, Rheinberger M, Verweij N, Bouatia-Naji N, Martin NG, Hastie N, Probst-Hensch N, Soranzo N, Devuyst O, Raitakari O, Gottesman O, Franco OH, Polasek O, Gasparini P, Munroe PB, Ridker PM, Mitchell P, Muntner P, Meisinger C, Smit JH, Kovacs P, Wild PS, Froguel P, Rettig R, Mägi R, Biffar R, Schmidt R, Middelberg RP, Carroll RJ, Penninx BW, Scott RJ, Katz R, Sedaghat S, Wild SH, Kardia SL, Ulivi S, Hwang SJ, Enroth S, Kloiber S, Trompet S, Stengel B, Hancock SJ, Turner ST, Rosas SE, Stracke S, Harris TB, Zeller T, Zemunik T, Lehtimäki T, Illig T, Aspelund T, Nikopensius T, Esko T, Tanaka T, Gyllensten U, Völker U, Emilsson V, Vitart V, Aalto V, Gudnason V, Chouraki V, Chen WM, Igl W, März W, Koenig W, Lieb W, Loos RJ, Liu Y, Snieder H, Pramstaller PP, Parsa A, O’Connell JR, Susztak K, Hamet P, Tremblay J, de Boer IH, Böger CA, Goessling W, Chasman DI, Köttgen A, Kao WH, Fox CS; ICBP Consortium; AGEN Consortium; CARDIOGRAM; CHARGe-Heart Failure Group; ECHOGen Consortium: Genetic associations at 53 loci highlight cell types and biological pathways relevant for kidney function. Nat Commun 7: 10023, 2016 26831199
5. Aulchenko YS, Ripatti S, Lindqvist I, Boomsma D, Heid IM, Pramstaller PP, Penninx BWJH, Janssens ACJW, Wilson JF, Spector T, Martin NG, Pedersen NL, Kyvik KO, Kaprio J, Hofman A, Freimer NB, Jarvelin M-R, Gyllensten U, Campbell H, Rudan I, Johansson A, Marroni F, Hayward C, Vitart V, Jonasson I, Pattaro C, Wright A, Hastie N, Pichler I, Hicks AA, Falchi M, Willemsen G, Hottenga J-J, de Geus EJC, Montgomery GW, Whitfield J, Magnusson P, Saharinen J, Perola M, Silander K, Isaacs A, Sijbrands EJG, Uitterlinden AG, Witteman JCM, Oostra BA, Elliott P, Ruokonen A, Sabatti C, Gieger C, Meitinger T, Kronenberg F, Döring A, Wichmann H-E, Smit JH, McCarthy MI, van Duijn CM, Peltonen L; ENGAGE Consortium: Loci influencing lipid levels and coronary heart disease risk in 16 European population cohorts. Nat Genet 41: 47–55, 2009 19060911
6. Teslovich TM, Musunuru K, Smith AV, Edmondson AC, Stylianou IM, Koseki M, Pirruccello JP, Ripatti S, Chasman DI, Willer CJ, Johansen CT, Fouchier SW, Isaacs A, Peloso GM, Barbalic M, Ricketts SL, Bis JC, Aulchenko YS, Thorleifsson G, Feitosa MF, Chambers J, Orho-Melander M, Melander O, Johnson T, Li X, Guo X, Li M, Shin Cho Y, Jin Go M, Jin Kim Y, Lee J-Y, Park T, Kim K, Sim X, Twee-Hee Ong R, Croteau-Chonka DC, Lange LA, Smith JD, Song K, Hua Zhao J, Yuan X, Luan J, Lamina C, Ziegler A, Zhang W, Zee RY, Wright AF, Witteman JC, Wilson JF, Willemsen G, Wichmann HE, Whitfield JB, Waterworth DM, Wareham NJ, Waeber G, Vollenweider P, Voight BF, Vitart V, Uitterlinden AG, Uda M, Tuomilehto J, Thompson JR, Tanaka T, Surakka I, Stringham HM, Spector TD, Soranzo N, Smit JH, Sinisalo J, Silander K, Sijbrands EJ, Scuteri A, Scott J, Schlessinger D, Sanna S, Salomaa V, Saharinen J, Sabatti C, Ruokonen A, Rudan I, Rose LM, Roberts R, Rieder M, Psaty BM, Pramstaller PP, Pichler I, Perola M, Penninx BW, Pedersen NL, Pattaro C, Parker AN, Pare G, Oostra BA, O’Donnell CJ, Nieminen MS, Nickerson DA, Montgomery GW, Meitinger T, McPherson R, McCarthy MI, McArdle W, Masson D, Martin NG, Marroni F, Mangino M, Magnusson PK, Lucas G, Luben R, Loos RJ, Lokki ML, Lettre G, Langenberg C, Launer LJ, Lakatta EG, Laaksonen R, Kyvik KO, Kronenberg F, König IR, Khaw KT, Kaprio J, Kaplan LM, Johansson A, Jarvelin MR, Janssens AC, Ingelsson E, Igl W, Kees Hovingh G, Hottenga JJ, Hofman A, Hicks AA, Hengstenberg C, Heid IM, Hayward C, Havulinna AS, Hastie ND, Harris TB, Haritunians T, Hall AS, Gyllensten U, Guiducci C, Groop LC, Gonzalez E, Gieger C, Freimer NB, Ferrucci L, Erdmann J, Elliott P, Ejebe KG, Döring A, Dominiczak AF, Demissie S, Deloukas P, de Geus EJ, de Faire U, Crawford G, Collins FS, Chen YD, Caulfield MJ, Campbell H, Burtt NP, Bonnycastle LL, Boomsma DI, Boekholdt SM, Bergman RN, Barroso I, Bandinelli S, Ballantyne CM, Assimes TL, Quertermous T, Altshuler D, Seielstad M, Wong TY, Tai ES, Feranil AB, Kuzawa CW, Adair LS, Taylor HA Jr., Borecki IB, Gabriel SB, Wilson JG, Holm H, Thorsteinsdottir U, Gudnason V, Krauss RM, Mohlke KL, Ordovas JM, Munroe PB, Kooner JS, Tall AR, Hegele RA, Kastelein JJ, Schadt EE, Rotter JI, Boerwinkle E, Strachan DP, Mooser V, Stefansson K, Reilly MP, Samani NJ, Schunkert H, Cupples LA, Sandhu MS, Ridker PM, Rader DJ, van Duijn CM, Peltonen L, Abecasis GR, Boehnke M, Kathiresan S: Biological, clinical and population relevance of 95 loci for blood lipids. Nature 466: 707–713, 2010 20686565
7. Ehret GB, Munroe PB, Rice KM, Bochud M, Johnson AD, Chasman DI, Smith AV, Tobin MD, Verwoert GC, Hwang S-J, Pihur V, Vollenweider P, O’Reilly PF, Amin N, Bragg-Gresham JL, Teumer A, Glazer NL, Launer L, Zhao JH, Aulchenko Y, Heath S, Sõber S, Parsa A, Luan J, Arora P, Dehghan A, Zhang F, Lucas G, Hicks AA, Jackson AU, Peden JF, Tanaka T, Wild SH, Rudan I, Igl W, Milaneschi Y, Parker AN, Fava C, Chambers JC, Fox ER, Kumari M, Go MJ, van der Harst P, Kao WH, Sjögren M, Vinay DG, Alexander M, Tabara Y, Shaw-Hawkins S, Whincup PH, Liu Y, Shi G, Kuusisto J, Tayo B, Seielstad M, Sim X, Nguyen KD, Lehtimäki T, Matullo G, Wu Y, Gaunt TR, Onland-Moret NC, Cooper MN, Platou CG, Org E, Hardy R, Dahgam S, Palmen J, Vitart V, Braund PS, Kuznetsova T, Uiterwaal CS, Adeyemo A, Palmas W, Campbell H, Ludwig B, Tomaszewski M, Tzoulaki I, Palmer ND, Aspelund T, Garcia M, Chang YP, O’Connell JR, Steinle NI, Grobbee DE, Arking DE, Kardia SL, Morrison AC, Hernandez D, Najjar S, McArdle WL, Hadley D, Brown MJ, Connell JM, Hingorani AD, Day IN, Lawlor DA, Beilby JP, Lawrence RW, Clarke R, Hopewell JC, Ongen H, Dreisbach AW, Li Y, Young JH, Bis JC, Kähönen M, Viikari J, Adair LS, Lee NR, Chen MH, Olden M, Pattaro C, Bolton JA, Köttgen A, Bergmann S, Mooser V, Chaturvedi N, Frayling TM, Islam M, Jafar TH, Erdmann J, Kulkarni SR, Bornstein SR, Grässler J, Groop L, Voight BF, Kettunen J, Howard P, Taylor A, Guarrera S, Ricceri F, Emilsson V, Plump A, Barroso I, Khaw KT, Weder AB, Hunt SC, Sun YV, Bergman RN, Collins FS, Bonnycastle LL, Scott LJ, Stringham HM, Peltonen L, Perola M, Vartiainen E, Brand SM, Staessen JA, Wang TJ, Burton PR, Soler Artigas M, Dong Y, Snieder H, Wang X, Zhu H, Lohman KK, Rudock ME, Heckbert SR, Smith NL, Wiggins KL, Doumatey A, Shriner D, Veldre G, Viigimaa M, Kinra S, Prabhakaran D, Tripathy V, Langefeld CD, Rosengren A, Thelle DS, Corsi AM, Singleton A, Forrester T, Hilton G, McKenzie CA, Salako T, Iwai N, Kita Y, Ogihara T, Ohkubo T, Okamura T, Ueshima H, Umemura S, Eyheramendy S, Meitinger T, Wichmann HE, Cho YS, Kim HL, Lee JY, Scott J, Sehmi JS, Zhang W, Hedblad B, Nilsson P, Smith GD, Wong A, Narisu N, Stančáková A, Raffel LJ, Yao J, Kathiresan S, O’Donnell CJ, Schwartz SM, Ikram MA, Longstreth WT Jr., Mosley TH, Seshadri S, Shrine NR, Wain LV, Morken MA, Swift AJ, Laitinen J, Prokopenko I, Zitting P, Cooper JA, Humphries SE, Danesh J, Rasheed A, Goel A, Hamsten A, Watkins H, Bakker SJ, van Gilst WH, Janipalli CS, Mani KR, Yajnik CS, Hofman A, Mattace-Raso FU, Oostra BA, Demirkan A, Isaacs A, Rivadeneira F, Lakatta EG, Orru M, Scuteri A, Ala-Korpela M, Kangas AJ, Lyytikäinen LP, Soininen P, Tukiainen T, Würtz P, Ong RT, Dörr M, Kroemer HK, Völker U, Völzke H, Galan P, Hercberg S, Lathrop M, Zelenika D, Deloukas P, Mangino M, Spector TD, Zhai G, Meschia JF, Nalls MA, Sharma P, Terzic J, Kumar MV, Denniff M, Zukowska-Szczechowska E, Wagenknecht LE, Fowkes FG, Charchar FJ, Schwarz PE, Hayward C, Guo X, Rotimi C, Bots ML, Brand E, Samani NJ, Polasek O, Talmud PJ, Nyberg F, Kuh D, Laan M, Hveem K, Palmer LJ, van der Schouw YT, Casas JP, Mohlke KL, Vineis P, Raitakari O, Ganesh SK, Wong TY, Tai ES, Cooper RS, Laakso M, Rao DC, Harris TB, Morris RW, Dominiczak AF, Kivimaki M, Marmot MG, Miki T, Saleheen D, Chandak GR, Coresh J, Navis G, Salomaa V, Han BG, Zhu X, Kooner JS, Melander O, Ridker PM, Bandinelli S, Gyllensten UB, Wright AF, Wilson JF, Ferrucci L, Farrall M, Tuomilehto J, Pramstaller PP, Elosua R, Soranzo N, Sijbrands EJ, Altshuler D, Loos RJ, Shuldiner AR, Gieger C, Meneton P, Uitterlinden AG, Wareham NJ, Gudnason V, Rotter JI, Rettig R, Uda M, Strachan DP, Witteman JC, Hartikainen AL, Beckmann JS, Boerwinkle E, Vasan RS, Boehnke M, Larson MG, Järvelin MR, Psaty BM, Abecasis GR, Chakravarti A, Elliott P, van Duijn CM, Newton-Cheh C, Levy D, Caulfield MJ, Johnson T; International Consortium for Blood Pressure Genome-Wide Association Studies; CARDIoGRAM Consortium; CKDGen Consortium; KidneyGen Consortium; EchoGen consortium; CHARGE-HF consortium: Genetic variants in novel pathways influence blood pressure and cardiovascular disease risk. Nature 478: 103–109, 2011 21909115
8. Yang J, Ferreira T, Morris AP, Medland SE, Madden PA, Heath AC, Martin NG, Montgomery GW, Weedon MN, Loos RJ, Frayling TM, McCarthy MI, Hirschhorn JN, Goddard ME, Visscher PM; Genetic Investigation of ANthropometric Traits (GIANT) Consortium; DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) Consortium: Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits. Nat Genet 44: 369–375, S1–S3, 2012 22426310
9. Mahajan A, Go MJ, Zhang W, Below JE, Gaulton KJ, Ferreira T, Horikoshi M, Johnson AD, Ng MC, Prokopenko I, Saleheen D, Wang X, Zeggini E, Abecasis GR, Adair LS, Almgren P, Atalay M, Aung T, Baldassarre D, Balkau B, Bao Y, Barnett AH, Barroso I, Basit A, Been LF, Beilby J, Bell GI, Benediktsson R, Bergman RN, Boehm BO, Boerwinkle E, Bonnycastle LL, Burtt N, Cai Q, Campbell H, Carey J, Cauchi S, Caulfield M, Chan JC, Chang LC, Chang TJ, Chang YC, Charpentier G, Chen CH, Chen H, Chen YT, Chia KS, Chidambaram M, Chines PS, Cho NH, Cho YM, Chuang LM, Collins FS, Cornelis MC, Couper DJ, Crenshaw AT, van Dam RM, Danesh J, Das D, de Faire U, Dedoussis G, Deloukas P, Dimas AS, Dina C, Doney AS, Donnelly PJ, Dorkhan M, van Duijn C, Dupuis J, Edkins S, Elliott P, Emilsson V, Erbel R, Eriksson JG, Escobedo J, Esko T, Eury E, Florez JC, Fontanillas P, Forouhi NG, Forsen T, Fox C, Fraser RM, Frayling TM, Froguel P, Frossard P, Gao Y, Gertow K, Gieger C, Gigante B, Grallert H, Grant GB, Grrop LC, Groves CJ, Grundberg E, Guiducci C, Hamsten A, Han BG, Hara K, Hassanali N, Hattersley AT, Hayward C, Hedman AK, Herder C, Hofman A, Holmen OL, Hovingh K, Hreidarsson AB, Hu C, Hu FB, Hui J, Humphries SE, Hunt SE, Hunter DJ, Hveem K, Hydrie ZI, Ikegami H, Illig T, Ingelsson E, Islam M, Isomaa B, Jackson AU, Jafar T, James A, Jia W, Jöckel KH, Jonsson A, Jowett JB, Kadowaki T, Kang HM, Kanoni S, Kao WH, Kathiresan S, Kato N, Katulanda P, Keinanen-Kiukaanniemi KM, Kelly AM, Khan H, Khaw KT, Khor CC, Kim HL, Kim S, Kim YJ, Kinnunen L, Klopp N, Kong A, Korpi-Hyövälti E, Kowlessur S, Kraft P, Kravic J, Kristensen MM, Krithika S, Kumar A, Kumate J, Kuusisto J, Kwak SH, Laakso M, Lagou V, Lakka TA, Langenberg C, Langford C, Lawrence R, Leander K, Lee JM, Lee NR, Li M, Li X, Li Y, Liang J, Liju S, Lim WY, Lind L, Lindgren CM, Lindholm E, Liu CT, Liu JJ, Lobbens S, Long J, Loos RJ, Lu W, Luan J, Lyssenko V, Ma RC, Maeda S, Mägi R, Männisto S, Matthews DR, Meigs JB, Melander O, Metspalu A, Meyer J, Mirza G, Mihailov E, Moebus S, Mohan V, Mohlke KL, Morris AD, Mühleisen TW, Müller-Nurasyid M, Musk B, Nakamura J, Nakashima E, Navarro P, Ng PK, Nica AC, Nilsson PM, Njølstad I, Nöthen MM, Ohnaka K, Ong TH, Owen KR, Palmer CN, Pankow JS, Park KS, Parkin M, Pechlivanis S, Pedersen NL, Peltonen L, Perry JR, Peters A, Pinidiyapathirage JM, Platou CG, Potter S, Price JF, Qi L, Radha V, Rallidis L, Rasheed A, Rathman W, Rauramaa R, Raychaudhuri S, Rayner NW, Rees SD, Rehnberg E, Ripatti S, Robertson N, Roden M, Rossin EJ, Rudan I, Rybin D, Saaristo TE, Salomaa V, Saltevo J, Samuel M, Sanghera DK, Saramies J, Scott J, Scott LJ, Scott RA, Segrè AV, Sehmi J, Sennblad B, Shah N, Shah S, Shera AS, Shu XO, Shuldiner AR, Sigurđsson G, Sijbrands E, Silveira A, Sim X, Sivapalaratnam S, Small KS, So WY, Stančáková A, Stefansson K, Steinbach G, Steinthorsdottir V, Stirrups K, Strawbridge RJ, Stringham HM, Sun Q, Suo C, Syvänen AC, Takayanagi R, Takeuchi F, Tay WT, Teslovich TM, Thorand B, Thorleifsson G, Thorsteinsdottir U, Tikkanen E, Trakalo J, Tremoli E, Trip MD, Tsai FJ, Tuomi T, Tuomilehto J, Uitterlinden AG, Valladares-Salgado A, Vedantam S, Veglia F, Voight BF, Wang C, Wareham NJ, Wennauer R, Wickremasinghe AR, Wilsgaard T, Wilson JF, Wiltshire S, Winckler W, Wong TY, Wood AR, Wu JY, Wu Y, Yamamoto K, Yamauchi T, Yang M, Yengo L, Yokota M, Young R, Zabaneh D, Zhang F, Zhang R, Zheng W, Zimmet PZ, Altshuler D, Bowden DW, Cho YS, Cox NJ, Cruz M, Hanis CL, Kooner J, Lee JY, Seielstad M, Teo YY, Boehnke M, Parra EJ, Chambers JC, Tai ES, McCarthy MI, Morris AP; DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) Consortium; Asian Genetic Epidemiology Network Type 2 Diabetes (AGEN-T2D) Consortium; South Asian Type 2 Diabetes (SAT2D) Consortium; Mexican American Type 2 Diabetes (MAT2D) Consortium; Type 2 Diabetes Genetic Exploration by Nex-Generation Sequencing in Multi-Ethnic Samples (T2D-GENES) Consortium: Genome-wide trans-ancestry meta-analysis provides insight into the genetic architecture of type 2 diabetes susceptibility. Nat Genet 46: 234–244, 2014 24509480
10. Wuttke M, Li Y, Li M, Sieber KB, Feitosa MF, Gorski M, Tin A, Wang L, Chu AY, Hoppmann A, Kirsten H, Giri A, Chai J-F, Sveinbjornsson G, Tayo BO, Nutile T, Fuchsberger C, Marten J, Cocca M, Ghasemi S, Xu Y, Horn K, Noce D, van der Most PJ, Sedaghat S, Yu Z, Akiyama M, Afaq S, Ahluwalia TS, Almgren P, Amin N, Ärnlöv J, Bakker SJL, Bansal N, Baptista D, Bergmann S, Biggs ML, Biino G, Boehnke M, Boerwinkle E, Boissel M, Bottinger EP, Boutin TS, Brenner H, Brumat M, Burkhardt R, Butterworth AS, Campana E, Campbell A, Campbell H, Canouil M, Carroll RJ, Catamo E, Chambers JC, Chee M-L, Chee M-L, Chen X, Cheng C-Y, Cheng Y, Christensen K, Cifkova R, Ciullo M, Concas MP, Cook JP, Coresh J, Corre T, Sala CF, Cusi D, Danesh J, Daw EW, de Borst MH, De Grandi A, de Mutsert R, de Vries APJ, Degenhardt F, Delgado G, Demirkan A, Di Angelantonio E, Dittrich K, Divers J, Dorajoo R, Eckardt K-U, Ehret G, Elliott P, Endlich K, Evans MK, Felix JF, Foo VHX, Franco OH, Franke A, Freedman BI, Freitag-Wolf S, Friedlander Y, Froguel P, Gansevoort RT, Gao H, Gasparini P, Gaziano JM, Giedraitis V, Gieger C, Girotto G, Giulianini F, Gögele M, Gordon SD, Gudbjartsson DF, Gudnason V, Haller T, Hamet P, Harris TB, Hartman CA, Hayward C, Hellwege JN, Heng C-K, Hicks AA, Hofer E, Huang W, Hutri-Kähönen N, Hwang S-J, Ikram MA, Indridason OS, Ingelsson E, Ising M, Jaddoe VWV, Jakobsdottir J, Jonas JB, Joshi PK, Josyula NS, Jung B, Kähönen M, Kamatani Y, Kammerer CM, Kanai M, Kastarinen M, Kerr SM, Khor C-C, Kiess W, Kleber ME, Koenig W, Kooner JS, Körner A, Kovacs P, Kraja AT, Krajcoviechova A, Kramer H, Krämer BK, Kronenberg F, Kubo M, Kühnel B, Kuokkanen M, Kuusisto J, La Bianca M, Laakso M, Lange LA, Langefeld CD, Lee JJ-M, Lehne B, Lehtimäki T, Lieb W, Lim SC, Lind L, Lindgren CM, Liu J, Liu J, Loeffler M, Loos RJF, Lucae S, Lukas MA, Lyytikäinen LP, Mägi R, Magnusson PKE, Mahajan A, Martin NG, Martins J, März W, Mascalzoni D, Matsuda K, Meisinger C, Meitinger T, Melander O, Metspalu A, Mikaelsdottir EK, Milaneschi Y, Miliku K, Mishra PP, Mohlke KL, Mononen N, Montgomery GW, Mook-Kanamori DO, Mychaleckyj JC, Nadkarni GN, Nalls MA, Nauck M, Nikus K, Ning B, Nolte IM, Noordam R, O’Connell J, O’Donoghue ML, Olafsson I, Oldehinkel AJ, Orho-Melander M, Ouwehand WH, Padmanabhan S, Palmer ND, Palsson R, Penninx BWJH, Perls T, Perola M, Pirastu M, Pirastu N, Pistis G, Podgornaia AI, Polasek O, Ponte B, Porteous DJ, Poulain T, Pramstaller PP, Preuss MH, Prins BP, Province MA, Rabelink TJ, Raffield LM, Raitakari OT, Reilly DF, Rettig R, Rheinberger M, Rice KM, Ridker PM, Rivadeneira F, Rizzi F, Roberts DJ, Robino A, Rossing P, Rudan I, Rueedi R, Ruggiero D, Ryan KA, Saba Y, Sabanayagam C, Salomaa V, Salvi E, Saum KU, Schmidt H, Schmidt R, Schöttker B, Schulz CA, Schupf N, Shaffer CM, Shi Y, Smith AV, Smith BH, Soranzo N, Spracklen CN, Strauch K, Stringham HM, Stumvoll M, Svensson PO, Szymczak S, Tai ES, Tajuddin SM, Tan NYQ, Taylor KD, Teren A, Tham YC, Thiery J, Thio CHL, Thomsen H, Thorleifsson G, Toniolo D, Tönjes A, Tremblay J, Tzoulaki I, Uitterlinden AG, Vaccargiu S, van Dam RM, van der Harst P, van Duijn CM, Velez Edward DR, Verweij N, Vogelezang S, Völker U, Vollenweider P, Waeber G, Waldenberger M, Wallentin L, Wang YX, Wang C, Waterworth DM, Bin Wei W, White H, Whitfield JB, Wild SH, Wilson JF, Wojczynski MK, Wong C, Wong TY, Xu L, Yang Q, Yasuda M, Yerges-Armstrong LM, Zhang W, Zonderman AB, Rotter JI, Bochud M, Psaty BM, Vitart V, Wilson JG, Dehghan A, Parsa A, Chasman DI, Ho K, Morris AP, Devuyst O, Akilesh S, Pendergrass SA, Sim X, Böger CA, Okada Y, Edwards TL, Snieder H, Stefansson K, Hung AM, Heid IM, Scholz M, Teumer A, Köttgen A, Pattaro C; Lifelines Cohort Study; V. A. Million Veteran Program: A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nat Genet 51: 957–972, 2019 31152163
11. Schizophrenia Working Group of the Psychiatric Genomics Consortium: Biological insights from 108 schizophrenia-associated genetic loci. Nature 511: 421–427, 2014
12. Köttgen A, Glazer NL, Dehghan A, Hwang S-J, Katz R, Li M, Yang Q, Gudnason V, Launer LJ, Harris TB, Smith AV, Arking DE, Astor BC, Boerwinkle E, Ehret GB, Ruczinski I, Scharpf RB, Chen Y-DI, de Boer IH, Haritunians T, Lumley T, Sarnak M, Siscovick D, Benjamin EJ, Levy D, Upadhyay A, Aulchenko YS, Hofman A, Rivadeneira F, Uitterlinden AG, van Duijn CM, Chasman DI, Paré G, Ridker PM, Kao WHL, Witteman JC, Coresh J, Shlipak MG, Fox CS: Multiple loci associated with indices of renal function and chronic kidney disease. Nat Genet 41: 712–717, 2009 19430482
13. Giral H, Landmesser U, Kratzer A: Into the wild: GWAS exploration of non-coding RNAs. Front Cardiovasc Med 5: 181, 2018 30619888
14. Zhang F, Lupski JR: Non-coding genetic variants in human disease. Hum Mol Genet 24: R102–R110, 2015 26152199
15. Musunuru K, Hickey KT, Al-Khatib SM, Delles C, Fornage M, Fox CS, Frazier L, Gelb BD, Herrington DM, Lanfear DE, Rosand J; American Heart Association Council on Functional Genomics and Translational Biology,Council on Clinical Cardiology, Council on Cardiovascular Disease in the Young, Council on Cardiovascular and Stroke Nursing, Council on Epidemiology and Prevention, Council on Hypertension, Council on Lifestyle and Cardiometabolic Health, Council on Quality of Care and Outcomes Research, and Stroke Council: Basic concepts and potential applications of genetics and genomics for cardiovascular and stroke clinicians: A scientific statement from the American Heart Association. Circ Cardiovasc Genet 8: 216–242, 2015 25561044
16. Höglund J, Rafati N, Rask-Andersen M, Enroth S, Karlsson T, Ek WE, Johansson Å: Improved power and precision with whole genome sequencing data in genome-wide association studies of inflammatory biomarkers. Sci Rep 9: 16844, 2019 31727947
17. Lin J, Musunuru K: From genotype to phenotype: A primer on the functional follow-up of genome-wide association studies in cardiovascular disease. Circ Genom Precis Med 11: e001946, 2018 29915816
18. Natarajan A, Yardimci GG, Sheffield NC, Crawford GE, Ohler U: Predicting cell-type-specific gene expression from regions of open chromatin. Genome Res 22: 1711–1722, 2012 22955983
19. Kiryluk K, Li Y, Sanna-Cherchi S, Rohanizadegan M, Suzuki H, Eitner F, Snyder HJ, Choi M, Hou P, Scolari F, Izzi C, Gigante M, Gesualdo L, Savoldi S, Amoroso A, Cusi D, Zamboli P, Julian BA, Novak J, Wyatt RJ, Mucha K, Perola M, Kristiansson K, Viktorin A, Magnusson PK, Thorleifsson G, Thorsteinsdottir U, Stefansson K, Boland A, Metzger M, Thibaudin L, Wanner C, Jager KJ, Goto S, Maixnerova D, Karnib HH, Nagy J, Panzer U, Xie J, Chen N, Tesar V, Narita I, Berthoux F, Floege J, Stengel B, Zhang H, Lifton RP, Gharavi AG: Geographic differences in genetic susceptibility to IgA nephropathy: GWAS replication study and geospatial risk analysis. PLoS Genet 8: e1002765, 2012 22737082
20. Genovese G, Friedman DJ, Ross MD, Lecordier L, Uzureau P, Freedman BI, Bowden DW, Langefeld CD, Oleksyk TK, Uscinski Knob AL, Bernhardy AJ, Hicks PJ, Nelson GW, Vanhollebeke B, Winkler CA, Kopp JB, Pays E, Pollak MR: Association of trypanolytic ApoL1 variants with kidney disease in African Americans. Science 329: 841–845, 2010 20647424
21. Lannon H, Shah SS, Dias L, Blackler D, Alper SL, Pollak MR, Friedman DJ: Apolipoprotein L1 (APOL1) risk variant toxicity depends on the haplotype background. Kidney Int 96: 1303–1307, 2019 31611067
22. Kruzel-Davila E, Wasser WG, Skorecki K: APOL1 nephropathy: A population genetics and evolutionary medicine detective story. Semin Nephrol 37: 490–507, 2017 29110756
23. Sampson MG, Robertson CC, Martini S, Mariani LH, Lemley KV, Gillies CE, Otto EA, Kopp JB, Randolph A, Vega-Warner V, Eichinger F, Nair V, Gipson DS, Cattran DC, Johnstone DB, O’Toole JF, Bagnasco SM, Song PX, Barisoni L, Troost JP, Kretzler M, Sedor JR; Nephrotic Syndrome Study Network: Integrative genomics identifies novel associations with APOL1 risk genotypes in black NEPTUNE subjects. J Am Soc Nephrol 27: 814–823, 2016 26150607
24. Manrai AK, Funke BH, Rehm HL, Olesen MS, Maron BA, Szolovits P, Margulies DM, Loscalzo J, Kohane IS: Genetic misdiagnoses and the potential for health disparities. N Engl J Med 375: 655–665, 2016 27532831
25. Varshney A, VanRenterghem H, Orchard P, Boyle AP, Stitzel ML, Ucar D, Parker SCJ: Cell specificity of human regulatory annotations and their genetic effects on gene expression. Genetics 211: 549–562, 2019 30593493
26. Schaid DJ, Chen W, Larson NB: From genome-wide associations to candidate causal variants by statistical fine-mapping. Nat Rev Genet 19: 491–504, 2018 29844615
27. Lu Q, Richardson B: DNaseI hypersensitivity analysis of chromatin structure. Methods Mol Biol 287: 77–86, 2004 15273405
28. Buenrostro JD, Giresi PG, Zaba LC, Chang HY, Greenleaf WJ: Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat Methods 10: 1213–1218, 2013 24097267
29. Heintzman ND, Stuart RK, Hon G, Fu Y, Ching CW, Hawkins RD, Barrera LO, Van Calcar S, Qu C, Ching KA, Wang W, Weng Z, Green RD, Crawford GE, Ren B: Distinct and predictive chromatin signatures of transcriptional promoters and enhancers in the human genome. Nat Genet 39: 311–318, 2007 17277777
30. Heintzman ND, Hon GC, Hawkins RD, Kheradpour P, Stark A, Harp LF, Ye Z, Lee LK, Stuart RK, Ching CW, Ching KA, Antosiewicz-Bourget JE, Liu H, Zhang X, Green RD, Lobanenkov VV, Stewart R, Thomson JA, Crawford GE, Kellis M, Ren B: Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459: 108–112, 2009 19295514
31. Kimura H: Histone modifications for human epigenome analysis. J Hum Genet 58: 439–445, 2013 23739122
32. Aldred SF, Collins PJ, Davis CA, Doyle F, Epstein CB, Frietze S, Kaul R, Lajoie BR, Landt SG, Lee B-K, Pauli F, Rosenbloom KR, Sabo P, Safi A, Sanyal A, Simon JM, Trinklein ND, Altshuler RC, Brown JB, Cheng C, Djebali S, Dong X, Ernst J, Giardine B, Greven M, Hardison RC, Harris RS, Herrero J, Hoffman MM, Iyer S, Kellis M, Kundaje A, Li Q, Lin X, Parker SCJ, Reddy TE, Schlesinger F, Thurman RE, Wang J, Whitfield TW, Wilder SP, Wu W, Xi HS, Yip KY, Zhuang J, Birney E, Dunham I, Gunter C, Snyder M, Pazin MJ, Lowdon RF, Dillon LAL, Adams LB, Kelly CJ, Zhang J, Wexler JR, Green ED, Good PJ, Feingold EA, Dekker J, Elnitski L, Gerstein M, Giddings MC, Hardison RC, Hubbard TJ, Kent WJ, Margulies EH, Myers RM, Snyder M, Stamatoyannopoulos JA, Tenenbaum SA, White KP, Risk BA, Gunawardena HP, Kuiper HC, Maier CW, Xie L, Bernstein BE, Shoresh N, Kheradpour P, Mikkelsen TS, Gillespie S, Goren A, Ram O, Zhang X, Wang L, Issner R, Coyne MJ, Durham T, Ku M, Truong T, Ward LD, Altshuler RC, Eaton ML, Merkel A, Dobin A, Lassmann T, Mortazavi A, Tanzer A, Lagarde J, Lin W, Xue C, Marinov GK, Khatun J, Williams BA, Zaleski C, Röder M, Kokocinski F, Abdelhamid RF, Alioto T, Antoshechkin I, Baer MT, Batut P, Bell I, Bell K, Chakrabortty S, Chen X, Chrast J, Curado J, Drenkow J, Dumais E, Dumais J, Duttagupta R, Fastuca M, Fejes-Toth K, Ferreira P, Foissac S, Fullwood MJ, Gao H, Gonzalez D, Gordon A, Gunawardena HP, Howald C, Jha S, Johnson R, Kapranov P, Kingswood C, Li G, Luo OJ, Park E, Preall JB, Presaud K, Ribeca P, Risk BA, Robyr D, Sammeth M, Sandhu KS, Schaeffer L, See L-H, Skancke J, Suzuki AM, Takahashi H, Tilgner H, Wang H, Wrobel J, Yu Y, Hayashizaki Y, Harrow J, Hubbard TJ, Reymond A, Antonarakis SE, Hannon GJ, Ruan Y, Wold B, Carninci P, Guigó R, Gingeras TR, Rosenbloom KR, Sloan CA, Learned K, Malladi VS, Wong MC, Barber GP, Cline MS, Dreszer TR, Heitner SG, Karolchik D, Kent WJ, Kirkup VM, Meyer LR, Long JC, Maddren M, Raney BJ, Furey TS, Song L, Grasfeder LL, Giresi PG, Lee B-K, Battenhouse A, Sheffield NC, Showers KA, London D, Bhinge AA, Shestak C, Schaner MR, Ki Kim S, Zhang ZZ, Mieczkowski PA, Mieczkowska JO, Liu Z, McDaniell RM, Ni Y, Rashid NU, Kim MJ, Adar S, Zhang Z, Wang T, Winter D, Keefe D, Iyer VR, Lieb JD, Crawford GE, Li G, Sandhu KS, Zheng M, Wang P, Shahab A, Ruan X, Myers RM, Pauli F, Gertz J, Vielmetter J, Partridge E, Trout D, Varley KE, Gasper C, Bansal A, Pepke S, Jain P, Amrhein H, Bowling KM, Anaya M, Cross MK, King B, Muratet MA, Newberry KM, McCue K, Nesmith AS, Fisher-Aylor KI, Pusey B, DeSalvo G, Parker SL, Balasubramanian S, Davis NS, Meadows SK, Eggleston T, Gunter C, Newberry JS, Levy SE, Absher DM, Wong WH, Blow MJ, Visel A, Pennachio LA, Elnitski L, Parker SCJ, Petrykowska HM, Aken B, Barrell D, Barson G, Berry A, Bignell A, Boychenko V, Bussotti G, Davidson C, Derrien T, Despacio-Reyes G, Diekhans M, Ezkurdia I, Frankish A, Gilbert J, Gonzalez JM, Griffiths E, Harte R, Hendrix DA, Hunt T, Jungreis I, Kay M, Khurana E, Leng J, Lin MF, Loveland J, Lu Z, Manthravadi D, Mariotti M, Mudge J, Mukherjee G, Notredame C, Pei B, Rodriguez JM, Saunders G, Sboner A, Searle S, Snow C, Steward C, Tapanari E, Tress ML, van Baren MJ, Walters N, Washietl S, Wilming L, Zadissa A, Zhang Z, Brent M, Haussler D, Valencia A, Hubbard TJ, Landt SG, Frietze S, Abyzov A, Addleman N, Alexander RP, Auerbach RK, Bettinger K, Bhardwaj N, Boyle AP, Cao AR, Cayting P, Charos A, Cheng Y, Cheng C, Eastman C, Euskirchen G, Fleming J, Grubert F, Habegger L, Hariharan M, Harmanci A, Iyengar S, Jin VX, Karczewski KJ, Kasowski M, Lacroute P, Lam H, Lamarre-Vincent N, Leng J, Lian J, Lindahl-Allen M, Min R, Miotto B, Monahan H, Moqtaderi Z, Mu XJ, O’Geen H, Ouyang Z, Patacsil D, Pei B, Raha D, Ramirez L, Reed B, Rozowsky J, Sboner A, Shi M, Sisu C, Slifer T, Witt H, Wu L, Xu X, Yan K-K, Yang X, Yip KY, Zhang Z, Struhl K, Weissman SM, Farnham PJ, Snyder M, Tenenbaum SA, Penalva LO, Doyle F, Karmakar S, Landt SG, Bhanvadia RR, Choudhury A, Domanus M, Ma L, Moran J, Patacsil D, Slifer T, Victorsen A, Yang X, Snyder M, White KP, Auer T, Centanin L, Eichenlaub M, Gruhl F, Heermann S, Hoeckendorf B, Inoue D, Kellner T, Kirchmaier S, Mueller C, Reinhardt R, Schertel L, Schneider S, Sinn R, Wittbrodt B, Wittbrodt J, Weng Z, Whitfield TW, Wang J, Collins PJ, Aldred SF, Trinklein ND, Partridge EC, Myers RM, Dekker J, Jain G, Lajoie BR, Sanyal A, Balasundaram G, Bates DL, Byron R, Canfield TK, Diegel MJ, Dunn D, Ebersol AK, Frum T, Garg K, Gist E, Hansen RS, Boatman L, Haugen E, Humbert R, Jain G, Johnson AK, Johnson EM, Kutyavin TV, Lajoie BR, Lee K, Lotakis D, Maurano MT, Neph SJ, Neri FV, Nguyen ED, Qu H, Reynolds AP, Roach V, Rynes E, Sabo P, Sanchez ME, Sandstrom RS, Sanyal A, Shafer AO, Stergachis AB, Thomas S, Vernot B, Vierstra J, Vong S, Wang H, Weaver MA, Yan Y, Zhang M, Akey JM, Bender M, Dorschner MO, Groudine M, MacCoss MJ, Navas P, Stamatoyannopoulos G, Dekker J, Beal K, Brazma A, Flicek P, Johnson N, Keefe D, Lukk M, Luscombe NM, Sobral D, Vaquerizas JM, Wilder SP, Batzoglou S, Sidow A, Hussami N, Kyriazopoulou-Panagiotopoulou S, Libbrecht MW, Schaub MA, Hardison RC, Miller W, Giardine B, Harris RS, Wu W, Bickel PJ, Banfai B, Boley NP, Brown JB, Huang H, Li Q, Li JJ, Noble WS, Bilmes JA, Buske OJ, Hoffman MM, Sahu AD, Kharchenko PV, Park PJ, Baker D, Taylor J, Iyer S, Dong X, Greven M, Lin X, Wang J, Xi HS, Zhuang J, Alexander RP, Balasubramanian S, Cheng C, Harmanci A, Lochovsky L, Min R, Mu XJ, Yan K-K, Yip KY; ENCODE Project Consortium: An integrated encyclopedia of DNA elements in the human genome. Nature 489: 57–74, 2012 22955616
33. Bujold D, Morais DAL, Gauthier C, Côté C, Caron M, Kwan T, Chen KC, Laperle J, Markovits AN, Pastinen T, Caron B, Veilleux A, Jacques P, Bourque G: The International Human Epigenome Consortium data portal. Cell Syst 3: 496–499.e2, 2016 27863956
34. Melnikov A, Murugan A, Zhang X, Tesileanu T, Wang L, Rogov P, Feizi S, Gnirke A, Callan CG Jr., Kinney JB, Kellis M, Lander ES, Mikkelsen TS: Systematic dissection and optimization of inducible enhancers in human cells using a massively parallel reporter assay. Nat Biotechnol 30: 271–277, 2012 22371084
35. Lin J, Musunuru K: Genome engineering tools for building cellular models of disease. FEBS J 283: 3222–3231, 2016 27218233
36. Higashijima Y, Hirano S, Nangaku M, Nureki O: Applications of the CRISPR-Cas9 system in kidney research. Kidney Int 92: 324–335, 2017 28433382
37. Battle A, Brown CD, Engelhardt BE, Montgomery SB; 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; Lead analysts; Laboratory, Data Analysis &Coordinating Center (LDACC); NIH program management; Biospecimen collection; Pathology; eQTL manuscript working group; Battle A, Brown CD, Engelhardt BE, Montgomery SB: Genetic effects on gene expression across human tissues [published correction appears in Nature 553: 530, 2018]. Nature 550: 204–213, 2017
38. Huckins LM, Dobbyn A, Ruderfer DM, Hoffman G, Wang W, Pardiñas AF, Rajagopal VM, Als TD, T Nguyen H, Girdhar K, Boocock J, Roussos P, Fromer M, Kramer R, Domenici E, Gamazon ER, Purcell S, Demontis D, Børglum AD, Walters JTR, O’Donovan MC, Sullivan P, Owen MJ, Devlin B, Sieberts SK, Cox NJ, Im HK, Sklar P, Stahl EA; CommonMind Consortium; Schizophrenia Working Group of the Psychiatric Genomics Consortium; iPSYCH-GEMS Schizophrenia Working Group: Gene expression imputation across multiple brain regions provides insights into schizophrenia risk. Nat Genet 51: 659–674, 2019 30911161
39. Nanda V, Wang T, Pjanic M, Liu B, Nguyen T, Matic LP, Hedin U, Koplev S, Ma L, Franzén O, Ruusalepp A, Schadt EE, Björkegren JLM, Montgomery SB, Snyder MP, Quertermous T, Leeper NJ, Miller CL: Functional regulatory mechanism of smooth muscle cell-restricted LMOD1 coronary artery disease locus. PLoS Genet 14: e1007755, 2018 30444878
40. Miller CL, Pjanic M, Wang T, Nguyen T, Cohain A, Lee JD, Perisic L, Hedin U, Kundu RK, Majmudar D, Kim JB, Wang O, Betsholtz C, Ruusalepp A, Franzén O, Assimes TL, Montgomery SB, Schadt EE, Björkegren JLM, Quertermous T: Integrative functional genomics identifies regulatory mechanisms at coronary artery disease loci. Nat Commun 7: 12092, 2016 27386823
41. Xu X, Eales JM, Akbarov A, Guo H, Becker L, Talavera D, Ashraf F, Nawaz J, Pramanik S, Bowes J, Jiang X, Dormer J, Denniff M, Antczak A, Szulinska M, Wise I, Prestes PR, Glyda M, Bogdanski P, Zukowska-Szczechowska E, Berzuini C, Woolf AS, Samani NJ, Charchar FJ, Tomaszewski M: Molecular insights into genome-wide association studies of chronic kidney disease-defining traits. Nat Commun 9: 4800, 2018 30467309
42. Qiu C, Huang S, Park J, Park Y, Ko Y-A, Seasock MJ, Bryer JS, Xu X-X, Song W-C, Palmer M, Hill J, Guarnieri P, Hawkins J, Boustany-Kari CM, Pullen SS, Brown CD, Susztak K: Renal compartment-specific genetic variation analyses identify new pathways in chronic kidney disease. Nat Med 24: 1721–1731, 2018 30275566
43. Al-Awqati Q, Oliver JA: Stem cells in the kidney. Kidney Int 61: 387–395, 2002 11849378
44. Park J, Shrestha R, Qiu C, Kondo A, Huang S, Werth M, Li M, Barasch J, Suszták K: Single-cell transcriptomics of the mouse kidney reveals potential cellular targets of kidney disease. Science 360: 758–763, 2018 29622724
45. Wilson PC, Wu H, Kirita Y, Uchimura K, Ledru N, Rennke HG, Welling PA, Waikar SS, Humphreys BD: The single-cell transcriptomic landscape of early human diabetic nephropathy. Proc Natl Acad Sci U S A 116: 19619–19625, 2019 31506348
46. Young MD, Mitchell TJ, Vieira Braga FA, Tran MGB, Stewart BJ, Ferdinand JR, Collord G, Botting RA, Popescu D-M, Loudon KW, Vento-Tormo R, Stephenson E, Cagan A, Farndon SJ, Del Castillo Velasco-Herrera M, Guzzo C, Richoz N, Mamanova L, Aho T, Armitage JN, Riddick ACP, Mushtaq I, Farrell S, Rampling D, Nicholson J, Filby A, Burge J, Lisgo S, Maxwell PH, Lindsay S, Warren AY, Stewart GD, Sebire N, Coleman N, Haniffa M, Teichmann SA, Clatworthy M, Behjati S: Single-cell transcriptomes from human kidneys reveal the cellular identity of renal tumors. Science 361: 594–599, 2018 30093597
47. Gillies CE, Putler R, Menon R, Otto E, Yasutake K, Nair V, Hoover P, Lieb D, Li S, Eddy S, Fermin D, McNulty MT, Hacohen N, Kiryluk K, Kretzler M, Wen X, Sampson MG; Nephrotic Syndrome Study Network (NEPTUNE): An eQTL landscape of kidney tissue in human nephrotic syndrome. Am J Hum Genet 103: 232–244, 2018 30057032
48. Zhang T, Choi J, Kovacs MA, Shi J, Xu M, Goldstein AM, Trower AJ, Bishop DT, Iles MM, Duffy DL, MacGregor S, Amundadottir LT, Law MH, Loftus SK, Pavan WJ, Brown KM; NISC Comparative Sequencing Program; Melanoma Meta-Analysis Consortium: Cell-type-specific eQTL of primary melanocytes facilitates identification of melanoma susceptibility genes. Genome Res 28: 1621–1635, 2018 30333196
49. Glastonbury CA, Couto Alves A, El-Sayed Moustafa JS, Small KS: Cell-type heterogeneity in adipose tissue is associated with complex traits and reveals disease-relevant cell-specific eQTLs. Am J Hum Genet 104: 1013–1024, 2019 31130283
50. Wang X, Tucker NR, Rizki G, Mills R, Krijger PH, de Wit E, Subramanian V, Bartell E, Nguyen X-X, Ye J, Leyton-Mange J, Dolmatova EV, van der Harst P, de Laat W, Ellinor PT, Newton-Cheh C, Milan DJ, Kellis M, Boyer LA: Discovery and validation of sub-threshold genome-wide association study loci using epigenomic signatures. eLife 5: e10557, 2016 27162171
51. Stueve TR, Marconett CN, Zhou B, Borok Z, Laird-Offringa IA: The importance of detailed epigenomic profiling of different cell types within organs. Epigenomics 8: 817–829, 2016 27305639
52. Sun BB, Maranville JC, Peters JE, Stacey D, Staley JR, Blackshaw J, Burgess S, Jiang T, Paige E, Surendran P, Oliver-Williams C, Kamat MA, Prins BP, Wilcox SK, Zimmerman ES, Chi A, Bansal N, Spain SL, Wood AM, Morrell NW, Bradley JR, Janjic N, Roberts DJ, Ouwehand WH, Todd JA, Soranzo N, Suhre K, Paul DS, Fox CS, Plenge RM, Danesh J, Runz H, Butterworth AS: Genomic atlas of the human plasma proteome. Nature 558: 73–79, 2018 29875488
53. Gate RE, Cheng CS, Aiden AP, Siba A, Tabaka M, Lituiev D, Machol I, Gordon MG, Subramaniam M, Shamim M, Hougen KL, Wortman I, Huang S-C, Durand NC, Feng T, De Jager PL, Chang HY, Aiden EL, Benoist C, Beer MA, Ye CJ, Regev A: Genetic determinants of co-accessible chromatin regions in activated T cells across humans. Nat Genet 50: 1140–1150, 2018 29988122
54. Kumasaka N, Knights AJ, Gaffney DJ: High-resolution genetic mapping of putative causal interactions between regions of open chromatin. Nat Genet 51: 128–137, 2019 30478436
55. Gallagher MD, Chen-Plotkin AS: The post-GWAS era: From association to function. Am J Hum Genet 102: 717–730, 2018 29727686
56. Deng W, Blobel GA: Detecting long-range enhancer-promoter interactions by quantitative chromosome conformation capture. Methods Mol Biol 1468: 51–62, 2017 27662870
57. Claussnitzer M, Dankel SN, Kim K-H, Quon G, Meuleman W, Haugen C, Glunk V, Sousa IS, Beaudry JL, Puviindran V, Abdennur NA, Liu J, Svensson P-A, Hsu Y-H, Drucker DJ, Mellgren G, Hui C-C, Hauner H, Kellis M: FTO obesity variant circuitry and adipocyte browning in humans. N Engl J Med 373: 895–907, 2015 26287746
58. Sieber KB, Batorsky A, Siebenthall K, Hudkins KL, Vierstra JD, Sullivan S, Sur A, McNulty M, Sandstrom R, Reynolds A, Bates D, Diegel M, Dunn D, Nelson J, Buckley M, Kaul R, Sampson MG, Himmelfarb J, Alpers CE, Waterworth D, Akilesh S: Integrated functional genomic analysis enables annotation of kidney genome-wide association study loci. J Am Soc Nephrol 30: 421–441, 2019 30760496
59. Ko Y-A, Yi H, Qiu C, Huang S, Park J, Ledo N, Köttgen A, Li H, Rader DJ, Pack MA, Brown CD, Susztak K: Genetic-variation-driven gene-expression changes highlight genes with important functions for kidney disease. Am J Hum Genet 100: 940–953, 2017 28575649
60. Böger CA, Gorski M, Li M, Hoffmann MM, Huang C, Yang Q, Teumer A, Krane V, O’Seaghdha CM, Kutalik Z, Wichmann HE, Haak T, Boes E, Coassin S, Coresh J, Kollerits B, Haun M, Paulweber B, Köttgen A, Li G, Shlipak MG, Powe N, Hwang SJ, Dehghan A, Rivadeneira F, Uitterlinden A, Hofman A, Beckmann JS, Krämer BK, Witteman J, Bochud M, Siscovick D, Rettig R, Kronenberg F, Wanner C, Thadhani RI, Heid IM, Fox CS, Kao WH; CKDGen Consortium: Association of eGFR-related loci identified by GWAS with incident CKD and ESRD. PLoS Genet 7: e1002292, 2011 21980298
61. Menon MC, Chuang PY, Li Z, Wei C, Zhang W, Luan Y, Yi Z, Xiong H, Woytovich C, Greene I, Overbey J, Rosales I, Bagiella E, Chen R, Ma M, Li L, Ding W, Djamali A, Saminego M, O’Connell PJ, Gallon L, Colvin R, Schroppel B, He JC, Murphy B: Intronic locus determines SHROOM3 expression and potentiates renal allograft fibrosis. J Clin Invest 125: 208–221, 2015 25437874
62. Prokop JW, Yeo NC, Ottmann C, Chhetri SB, Florus KL, Ross EJ, Sosonkina N, Link BA, Freedman BI, Coppola CJ, McDermott-Roe C, Leysen S, Milroy L-G, Meijer FA, Geurts AM, Rauscher FJ 3rd, Ramaker R, Flister MJ, Jacob HJ, Mendenhall EM, Lazar J: Characterization of coding/noncoding variants for SHROOM3 in patients with CKD. J Am Soc Nephrol 29: 1525–1535, 2018 29476007
63. Han J, Liu Y, Rao F, Nievergelt CM, O’Connor DT, Wang X, Liu L, Bu D, Liang Y, Wang F, Zhang L, Zhang H, Chen Y, Wang H: Common genetic variants of the human uromodulin gene regulate transcription and predict plasma uric acid levels. Kidney Int 83: 733–740, 2013 23344472
64. Trudu M, Janas S, Lanzani C, Debaix H, Schaeffer C, Ikehata M, Citterio L, Demaretz S, Trevisani F, Ristagno G, Glaudemans B, Laghmani K, Dell’Antonio G, Loffing J, Rastaldi MP, Manunta P, Devuyst O, Rampoldi L; SKIPOGH team: Common noncoding UMOD gene variants induce salt-sensitive hypertension and kidney damage by increasing uromodulin expression. Nat Med 19: 1655–1660, 2013 24185693
65. Bruggeman LA, Wu Z, Luo L, Madhavan SM, Konieczkowski M, Drawz PE, Thomas DB, Barisoni L, Sedor JR, O’Toole JF: APOL1-G0 or APOL1-G2 transgenic models develop preeclampsia but not kidney disease. J Am Soc Nephrol 27: 3600–3610, 2016
66. Beckerman P, Bi-Karchin J, Park ASD, Qiu C, Dummer PD, Soomro I, Boustany-Kari CM, Pullen SS, Miner JH, Hu C-AA, Rohacs T, Inoue K, Ishibe S, Saleem MA, Palmer MB, Cuervo AM, Kopp JB, Susztak K: Transgenic expression of human APOL1 risk variants in podocytes induces kidney disease in mice. Nat Med 23: 429–438, 2017 28218918
67. Khera AV, Chaffin M, Aragam KG, Haas ME, Roselli C, Choi SH, Natarajan P, Lander ES, Lubitz SA, Ellinor PT, Kathiresan S: Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat Genet 50: 1219–1224, 2018 30104762
68. Levin MG, Rader DJ: Polygenic risk scores and coronary artery disease: Ready for prime time? Circulation 141: 637–640, 2020 32091922
69. Richardson TG, Harrison S, Hemani G, Davey Smith G: An atlas of polygenic risk score associations to highlight putative causal relationships across the human phenome. eLife 8: e43657, 2019 30835202
70. Hoste L, Deiteren K, Pottel H, Callewaert N, Martens F: Routine serum creatinine measurements: How well do we perform? BMC Nephrol 16: 21, 2015 25803560
Keywords:

Kidney Genomics Series; Genome-Wide Association Study; Computational Biology; Genomics; Genetic Association Studies; Renal Insufficiency; Chronic; DNA

Copyright © 2020 by the American Society of Nephrology