Abstract
Most of the genetic basis of chronic liver disease remains undiscovered. To identify novel genetic loci that modulate the risk of liver injury, we performed genome-wide association studies (GWAS) on circulating levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP) and total bilirubin across 312,671 White British participants in the UK Biobank. We focused on variants associated with elevations in all four liver biochemistries at genome-wide significance (P<5x10−8) and that replicated using Mass General Brigham Biobank in 19,323 European ancestry individuals. We identified a genetic locus in mitochondrial glycerol-3-phosphate acyltransferase (GPAM rs10787429) associated with increased levels of ALT (P=1.4x10−30), AST (P=3.6x10−10), ALP (P=9.5x10−30) and total bilirubin (P=2.9x10−12). This common genetic variant was also associated with an allele dose-dependent risk of alcoholic liver disease (OR 1.34, P=2.6x10−5) and fatty liver disease (OR 1.18, P=5.8x10−4) by ICD-10 codes. We identified significant interactions between GPAM rs10787429 and elevated body mass index in association with ALT and AST (P-interaction=7.1x10−9 and 3.95x10−8, respectively), as well as between GPAM rs10787429 and weekly alcohol consumption in association with ALT, AST, and alcoholic liver disease (P-interaction=4.0x10−2, 1.6x10−2 and 1.3x10−2, respectively). Unlike previously described genetic variants that are associated with an increased risk of liver injury but confer a protective effect on circulating lipids, GPAM rs10787429 was associated with an increase in total cholesterol (P=2.0x10−17), LDL cholesterol (P=2.0x10−10), and HDL cholesterol (P=6.6x10−37). Single-cell RNA sequencing data demonstrated hepatocyte-predominant expression of GPAM in cells that co-express genes related to VLDL production (P=9.4x10−103). In conclusion, genetic variation in GPAM is associated with susceptibility to liver injury. GPAM may represent a new therapeutic target in chronic liver disease.
Keywords: glycerol-3-phosphate acyltransferase, genomic analysis, precision medicine
Chronic liver disease leads to over 40,000 deaths annually in the United States and represents an area of substantial unmet medical need (1). While environmental factors play a role in the development of chronic liver disease, prospective twin studies have demonstrated that up to half of the observed variation in hepatic fibrosis and steatosis is attributable to genetic factors (2). Elucidation of the genetic underpinnings of chronic liver disease may reveal new targets for therapeutic intervention; drug development targets with human genetic evidence of disease association are more likely to lead to FDA-approved drugs (3).
Hepatocellular injury is characterized by elevations in serum alanine (ALT) and aspartate (AST) aminotransferases, while cholestatic liver injury is associated with elevated serum alkaline phosphatase (ALP) and total bilirubin levels. A powerful approach for understanding the molecular basis of liver disease has been to perform genome-wide association studies (GWAS) of levels of circulating liver enzymes across large population samples (4-13). Combined GWAS of ALT and AST have previously revealed sequence variations associated with liver disease, such as PNPLA3 I148M (11) and HSD17B13 rs72613567 (13); this has catalyzed the development of new therapeutics targeting these genes for the treatment of chronic liver disease (14). A more recent large trans-ethnic GWAS of ALT and AST identified a missense variant in the gene encoding manganese 28 efflux transporter SLC30A10 (rs188273166) associated with liver disease and extrahepatic cholangiocarcinoma (15).
A limitation of combined GWAS of liver transaminases is that elevations in ALT or AST may be caused by kidney, heart or muscle damage (16), and therefore only a subset of identified loci are likely to be causally implicated in liver damage. In addition, in contrast to other metabolic disorders, such as obesity or type two diabetes, where hundreds of genetic risk loci have been identified (17), few genetic associations have been identified to date for chronic liver disease. This highlights the need for studies with increased sample size and the exploration of novel liver-related endophenotypes.
To further study genetic variants that may confer susceptibility to liver injury, we performed GWAS on circulating levels of ALT, AST, ALP, and total bilirubin across 312,671 unrelated White British participants in UK Biobank, a large population-based study. We focused on variants associated with elevations in all four liver biochemistries at genome-wide significance (P<5x10−8), hypothesizing that genetic variants associated with both elevated liver aminotransferases (ALT, AST) and elevated markers of cholestasis (ALP, total bilirubin) would be more likely to play a role in liver health. We replicated our findings in 19,323 unrelated European ancestry subjects from the Mass General Brigham Biobank, a hospital-based cohort. For variants that associated with elevated risk of liver injury across both cohorts, we also sought to determine whether the variants were associated with clinical liver disease phenotypes by ICD-10 codes, whether the variants demonstrated differential effect by other risk factors for liver disease (gene by environment interaction), and whether the variants were associated with circulating lipids. Finally, we analyzed single-cell RNA sequencing data from healthy human livers to determine the relative contribution of cell sub-populations to the expression of identified target genes.
EXPERIMENTAL PROCEDURES
Cohort descriptions and quality control
The UK Biobank is a population-based cohort consisting of 502,682 individuals (18). To minimize confounding due to population structure in our dataset, we restricted our analysis to include only subjects estimated to have British ancestry. We also excluded individuals with more genome-wide heterozygosity than expected, an excess of missing genotype calls, putative sex chromosome aneuploidy, and more than 10 third-degree relatives. We further removed at least one individual from each related pair with kinship coefficient >0.0625, giving preference to inclusion of patients with all-cause cirrhosis by ICD-10 codes. After quality control, 312,671 unrelated White British subjects were included in the analysis. We replicated our findings using 19,323 unrelated (kinship coefficient <0.0885) European ancestry samples from the Mass General Brigham Biobank, a hospital-based cohort of 43,534 individuals (19).
Phenotype data
In UK Biobank, blood biochemistry values were obtained for ALT (field 30620), AST (field 20650), ALP (field 30610), total bilirubin (field 30840), cholesterol (field 30690), LDL direct (field 30780), and HDL cholesterol (field 30760). In Mass General Brigham Biobank, blood biochemistry values for ALT, AST and ALP were obtained. We log-transformed ALT, AST, ALP and total bilirubin, resulting in approximately normal distributions. We defined a severe alcoholic liver disease phenotype by combining the following ICD-10 codes: K702 (alcoholic fibrosis and sclerosis of liver), K703 (alcoholic cirrhosis of liver), and K704 (alcoholic hepatic failure). For the all-cause cirrhosis phenotype, we combined the severe alcoholic liver disease ICD-10 codes with K740 (hepatic fibrosis), K741 (hepatic sclerosis), K742 (hepatic fibrosis with hepatic sclerosis), K746 (other and unspecified cirrhosis of the liver), K766 (portal hypertension), I850 (bleeding esophageal varices), I859 (esophageal varices), K717 (toxic liver disease with fibrosis and cirrhosis of liver), K721 (chronic hepatic failure), K729 (hepatic failure, unspecified), and K767 (hepatorenal syndrome). We also investigated a phenotype for all-cause fatty liver by combining K700 (alcoholic fatty liver), K701 (alcoholic hepatitis), K709 (alcoholic liver disease, unspecified), K760 (fatty liver, not elsewhere classified), K758 (other specified inflammatory liver diseases), and K759 (inflammatory liver disease, unspecified).
Genotype data
In UK Biobank, genotyping was performed using either the UK BiLEVE Axiom array or the UK Biobank Axiom array, then imputed into the Haplotype Reference Consortium (HRC) and UK10K + 1000 Genomes panels. We used genotype data from the UK Biobank dataset release version 2 and the hg19 human genome reference for all analyses in this study. For Mass General Brigham Biobank, genotyping was performed using the Illumina MEGA array, QC steps were conducted consistent with prior studies (19), and imputation was performed to the HRC using the Michigan Imputation Server. We filtered out variants with minor allele frequency < 0.01 and imputation quality < 0.5. Content overlap between the UK Biobank and Mass General Brigham Biobank genotyping arrays was between 74.3% and 81.3% per chromosome.
Genome-wide association analyses in UK Biobank
We performed GWAS using linear regression for ALT, AST, ALP and total bilirubin, adjusting for age, sex, body mass index (BMI), total number of medications taken by each participant, genotyping batch, and the first 10 principal components of genetic ancestry. Association analyses were performed using linear regression in PLINK 2.0 alpha (20). Linkage disequilibrium score regression (LDSC) was used to estimate heritability and test statistic inflation due to confounding by population substructure (21). GWAS summary statistics were uploaded to the FUnctional Mapping and Annotation of GWAS tool (FUMA; version 1.3.5) (22). We used a panel of 10,000 randomly selected unrelated UK Biobank participants, release 2b, as the reference population for calculating linkage disequilibrium (LD). For each GWAS, we identified independent genomic risk loci defined by a genome-wide significance value of P<5x10−8, an r2 threshold of 0.2, and a maximum distance between LD blocks to merge into a locus of < 250 kb. To define shared association signals across the four phenotypes, we identified SNPs within these blocks with r2 > 0.8 that were genome-wide significant and in common across the ALT, AST, ALP and total bilirubin GWAS. The lead independent SNPs that generated the common proxy SNP were identified from the ALT, AST, ALP and total bilirubin GWAS, and the SNP with the smallest p-value was kept. To correct for potential test statistic inflation, we also performed a sensitivity analysis where we inflated GWAS standard errors by the square root of the estimated intercept from the LD score regression.
Replication of top results from UK Biobank in the Mass General Brigham Biobank
We focused on variants associated with elevations in all four liver biochemistries in UK Biobank at genome-wide significance (P<5x10−8) for replication in Mass General Brigham Biobank in 19,323 unrelated European ancestry individuals. We examined the association of these genetic variants with ALT, AST, and ALP using linear regression, assuming an additive model, and adjusting for age, sex, BMI, and first ten principal components of ancestry.
Association analyses for binary liver disease phenotypes and additional quantitative traits
For replicated variants, we investigated the association with binary liver disease phenotypes by ICD-10 codes in UK Biobank. We used R 3.6.3 to perform logistic regression. To account for case-control imbalance for binary liver disease phenotypes and lower allele frequencies, all logistic regression results were analyzed using the Firth penalized likelihood approach. We also examined the association of genetic variants with circulating lipids (total cholesterol, direct LDL, and HDL) using linear regression. We assumed an additive model for all association analyses and adjusted all models for age, sex, BMI, total number of medications taken by each participant, genotyping batch, and first ten principal components of ancestry.
Sensitivity analyses for alcohol use and type 2 diabetes in UK Biobank
Given the known important role of alcohol intake on liver injury, we performed a sensitivity analysis on variants that replicated in Mass General Brigham Biobank, using self-reported number of alcoholic drinks consumed as a covariate. We calculated the average units of alcohol consumed per week for each participant in UK Biobank, assuming 2 units (16g) of pure alcohol in a pint of beer/cider; 1.5 units (12g) in a glass of red wine, champagne, white wine, fortified wine, and “other” alcohol drink; and 1 unit (8g) in a measure of spirits. For participants who reported consuming alcohol monthly rather than weekly, we multiplied by 0.23 to convert monthly alcohol consumption to weekly. Weekly intake values greater than 6 standard deviations from the mean were excluded, reflective of greater than 92 alcoholic drinks per week. We also performed a sensitivity analysis adjusting for the presence of type 2 diabetes, which was defined as self-report of type 2 diabetes, followed by a verbal interview with a trained nurse to confirm the diagnosis, or hospitalization for diabetes (ICD code E11).
Assessing for gene-environment interactions in UK Biobank
For genetic variants associated with elevated risk of liver injury in both UK Biobank and Mass General Brigham Biobank, we investigated the combined effects of the genetic variant and BMI on biomarkers of hepatocellular injury. We performed an interaction analysis by modeling the main effects of the genetic variant and BMI, as well as an interaction term, using an additive model. BMI was entered as a continuous variable in all analyses. To depict the interaction between genotype and BMI visually, participants were divided into four categories of BMI: lean (<25 kg/m2), overweight (25–30 kg/m2), obese (30–35 kg/m2) and very obese (>35 kg/m2). We performed a similar analysis to assess the interaction between the genetic variant and weekly alcohol consumption on markers of hepatocellular injury and alcoholic liver disease. We depicted the interaction between genotype and alcohol use by dividing participants into four categories: no alcohol use, low risk alcohol use (<21 units of alcohol/week for men, <14 units of alcohol/week for women), hazardous drinking (22-49 units of alcohol/week for men, 15-35 units of alcohol/week for women), and harmful drinking (>50 units of alcohol/week for men, >36 units of alcohol/week for women) (23).
Single-cell RNA sequencing
Merged single-cell RNA data from non-diseased human livers was acquired (24). We assessed the expression of target genes of interest in parenchymal and non-parenchymal liver cells, and in hepatocytes with or without a gene expression signature of VLDL production. This signature was defined by co-expression of more than three transcripts of APOB and MTTP. Expression of target genes of interest in VLDL-producing compared to non-VLDL producing hepatocytes was tested for statistical difference using Wilcoxon rank sum test. Data was analyzed using Seurat and R 3.6.3.
RESULTS
Genome-wide association study and replication results
We performed GWAS of 9.9 million genetic variants on circulating levels of ALT, AST, ALP and total bilirubin across 312,671 unrelated White British participants in UK Biobank. SNP-based heritability estimates (standard error) in our sample were 10.8% (0.8%) for ALT, 10.7% (1.0%) for AST, 19% (2.6%) for ALP, and 10.1% (3.2%) for total bilirubin. According to the estimated intercept (standard error) from a linkage disequilibrium (LD) score regression, some of the genomic inflation in test statistics for ALT, AST, and total bilirubin may be due to biases such as residual population stratification: 1.0774 (0.013) for ALT; 1.0688 (0.01) for AST; 1.14 (0.0257) for ALP, and 1.0489 (0.0154) for total bilirubin. We identified 204 distinct genomic risk loci associated with ALT, 201 associated with AST, 274 associated with ALP, and 99 associated with total bilirubin at P<5x10−8. We next identified shared association signals across the four liver biomarkers, with traits having either the same index variant or a variant in strong linkage disequilibrium (r2 > 0.8 with the index variant). This identified 58 distinct loci associated with both ALT and AST, 18 distinct loci associated with both ALP and total bilirubin, and 5 distinct loci associated with ALT, AST, ALP and total bilirubin (Figure 1, Figure 2, Table 1). Of these 5 loci, 2 of them, rs10787429 (closest gene, GPAM) and rs11601507 (closest gene, TRIM5), were associated with elevated levels across all four liver biochemistries. The other 3 genetic variants, such as the well-characterized locus at SUGP1/TM6SF2 rs200210321, were highly associated with elevated ALT, AST and total bilirubin but were also associated with significantly decreased ALP (Table 1). We focused on the 2 identified variants associated with elevations across all four liver biochemistries, suggestive of a possible common pathway towards liver cholestasis and hepatocellular injury. We attempted to replicate the association of GPAM rs10787429 and TRIM5 rs11601507 with elevated markers of liver injury in Mass General Brigham Biobank, a hospital-based cohort. GPAM rs10787429 was associated with log-transformed ALT (P=2.7x10−3), AST (P=3.3x10−2) and ALP (P=1.9x10−2) in Mass General Brigham Biobank (Table 2). TRIM5 rs11601507 was not significantly associated with liver injury in Mass General Brigham Biobank, perhaps due to insufficient statistical power (minor allele frequency for GPAM rs10787429 0.234 compared to 0.06 for TRIM5 rs11601507) (Table 2). As GPAM rs10787429 was the only risk locus associated with elevated ALT, AST, ALP and total bilirubin in UK Biobank that replicated in Mass General Brigham Biobank, we focused further analysis on this locus (Figure 3). In UK Biobank, the results for GPAM rs10787429 remained at genome-wide significance after inflating the standard errors by the square root of the LD score regression intercept for each biomarker of liver injury GWAS (Supplementary Table 1).
Figure 1:
Manhattan plots of GWAS results in 312,671 unrelated White British participants in UK Biobank for (A) ALT, (B) AST. Red dots indicate lead SNPs for shared signals across all four liver biochemistry GWAS. Dark blue dots (A, B) indicate lead SNPs for shared loci between ALT and AST GWAS; the shared signals are marked only once, on the plot for which the p-value for the association is smaller. Dashed line indicates P<5x10−8.
Figure 2:
Manhattan plots of GWAS results in 312,671 unrelated White British participants in UK Biobank for (A) ALP, (B) total bilirubin. Red dots indicate lead SNPs for shared signals across all four liver biochemistry GWAS. Purple dots (A, B) indicate lead SNPs for shared loci between ALP and total bilirubin GWAS; the shared signals are marked only once, on the plot for which the p-value for the association is smaller. Dashed line indicates P<5x10−8.
Table 1:
Association signals shared across ALT, AST, ALP and total bilirubin GWAS studies, defined by having a lead SNP from each GWAS (ALT, AST, ALP, total bilirubin) either identical or with shared proxies with r2 > 0.8. The lead SNP with the smallest p-value was kept. Linear regression was performed adjusting for age, sex, BMI, number of medications, genotyping batch, and the first ten principal components of ancestry. Biomarkers of liver injury are log-transformed.
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
|---|---|---|---|---|
| GPAM rs10787429 | ALT | 1.37e−02 | 1.19e−03 | 1.37e−30 |
| [10:113949664:C:T] | AST | 4.84e−03 | 7.73e−04 | 3.59e−10 |
| ALP | 8.90e−03 | 7.85e−04 | 9.51e−30 | |
| total bilirubin | 7.56e−03 | 1.08e−03 | 2.93e−12 | |
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
| TRIM5 rs11601507 | ALT | 1.75e−02 | 2.08e−03 | 4.09e−17 |
| [11:5701074:A:C] | AST | 1.03e−02 | 1.35e−03 | 1.96e−14 |
| ALP | 1.98e−02 | 1.37e−03 | 3.23e−47 | |
| total bilirubin | 1.29e−02 | 1.89e−03 | 7.58e−12 | |
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
| NLRP3 rs56015600 | ALT | 7.92e−03 | 1.10e−03 | 5.51e−13 |
| [1:247602968:A:G] | AST | 3.55e−03 | 7.12e−04 | 6.33e−07 |
| ALP | −6.29e−03 | 7.24e−04 | 3.75e−18 | |
| total bilirubin | 6.37e−03 | 9.98e−04 | 1.68e−10 | |
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
| IL1F10 rs6743171 | ALT | −7.23e−03 | 1.09e−03 | 2.90e−11 |
| [2:113840058:C:G] | AST | −5.33e−03 | 7.04e−04 | 3.99e−14 |
| ALP | 6.01e−03 | 7.16e−04 | 4.74e−17 | |
| total bilirubin | −8.18e−03 | 9.87e−04 | 1.12e−16 | |
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
| SUGP1/TM6SF2 rs200210321 | ALT | 4.11e−02 | 2.04e−03 | 2.31e−90 |
| [19:19393890:A:AG] | AST | 1.89e−02 | 1.32e−03 | 1.93e−46 |
| ALP | −2.38e−02 | 1.34e−03 | 1.11e−70 | |
| total bilirubin | 1.47e−02 | 1.85e−03 | 2.12e−15 |
Table 2:
Association of genetic variants of interest with log-transformed biomarkers of liver injury in Mass General Brigham Biobank (n = 19,323). Variants were selected based on their association associated with elevated ALT, AST, ALP and total bilirubin in UK Biobank. Linear regression was performed adjusting for age, sex, BMI, batch, and the first ten principal components of ancestry.
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
|---|---|---|---|---|
| GPAM rs10787429 | ALT | 2.28e−02 | 7.60e−03 | 2.73e−03 |
| [10:113949664:C:T] | AST | 1.19e−02 | 5.59e−03 | 3.31e−02 |
| ALP | 1.32e−02 | 5.61e−03 | 1.87e−02 | |
| Variant | Measure (U/L) | Estimate | Std. Error | P-value |
| TRIM5 rs11601507 | ALT | 1.64e−02 | 1.31e−02 | 2.10e−01 |
| [11:5701074:A:C] | AST | 7.54e−03 | 9.63e−03 | 4.33e−01 |
| ALP | 1.85e−02 | 9.66e−03 | 5.57e−02 |
Figure 3:
Representative regional plot of GWAS p-values with genes and functional annotations of genomic risk loci on chromosome 10 for GWAS of ALT. Genes prioritized by FUMA are highlighted in red; SNPs are colored based on r2.
Association with ICD-10 based definitions of chronic liver disease in UK Biobank
We investigated the association of GPAM rs10787429 with broad categories of liver disease defined by ICD-10 codes in UK Biobank, including severe alcoholic liver disease (N=413), all-cause cirrhosis (N=1,493), and fatty liver disease (N=979). A Bonferroni adjusted significance level of P<0.017 was used for the three clinical liver disease phenotypes. GPAM rs10787429 was associated with increased risk of severe alcoholic liver disease (OR 1.34, P=2.6x10−5) and fatty liver disease (OR 1.18, P=5.8x10−4) in an allele dose-dependent manner; these odds were comparable to those of well-established genetic variants, including PNPLA3 I148M and TM6SF2 E167K (Figure 4). A nominally significant increased risk of all-cause cirrhosis was observed for GPAM rs10787429 (OR 1.09, P=2.6x10−2) (Supplementary Figure 1).
Figure 4:

Association of GPAM rs10787429, PNPLA3 I148M, and TM6SF2 E167K with (A) severe alcoholic liver disease and (B) all-cause fatty liver disease, by ICD-10 diagnostic codes in UK Biobank, using the Firth penalized likelihood approach. Odds ratios were calculated with the use of logistic regression, with adjustment for age, sex, BMI, total number of medications, genotyping batch, and first ten principal components of ancestry.
Sensitivity analyses for alcohol use and type 2 diabetes in UK Biobank
Given the role of alcohol intake on liver damage, we performed a sensitivity analysis analyzing the association of GPAM rs10787429 with biomarkers of liver damage while accounting for weekly alcohol consumption. The results were consistent and remained at genome-wide significance across all biomarkers of liver injury, suggesting GPAM rs10787429 has impact on liver damage independent of alcohol consumption (Supplementary Table 2). The associations also remained significant for alcoholic liver disease and fatty liver disease phenotypes (Supplementary Figure 2, 3). The results adjusted for the presence of type 2 diabetes were similar (Supplementary Table 3; Supplementary Figure 4, 5).
Analysis of gene-environment interactions in UK Biobank
To determine if the effect of the GPAM rs10787429 variant on hepatocellular injury is modified by BMI, we analyzed the relationship between GPAM rs10787429 and ALT and AST after stratifying UK Biobank participants into four categories based on BMI. We performed linear regression modelling main effects of GPAM rs10787429 and BMI as well as an interaction term, assuming an additive model. Significant interactions were identified with elevated BMI and GPAM rs10787429 in association with ALT (P-interaction=7.1x10−9) and AST (P-interaction=4.0x10−8), suggesting a gene-environment interaction (Figure 5). We also performed a similar analysis dividing participants into four categories of alcohol use: no alcohol use, low risk use, hazardous drinking, and harmful drinking, according to UK guidelines (23). We demonstrated a nominally significant interaction between elevated weekly alcohol use and GPAM rs10787429 in association with ALT (P-interaction=4.0x10−2) and AST (P-interaction=1.6x10−2), using self-reported weekly alcohol use as a continuous variable (Supplementary Figure 6). There was also a nominally significant interaction between alcohol consumption and GPAM rs10787429 in association with severe alcoholic liver disease (P-interaction=1.3x10−2).
Figure 5:
Alanine aminotransferase (ALT) and aspartate aminotransferase (AST) by BMI and GPAM rs10787429 status in UK Biobank. Circles and bars depict medians the interquartile ranges, respectively. The ALT- and AST- increasing effect of the GPAM rs10787429 was amplified by increasing BMI (p for interaction rs10787429 x BMI on ALT = 7.1x10−9; p for interaction rs10787429 x BMI on AST = 4.0x10−8).
Association with circulating lipids in UK Biobank
The previously described genetic variants PNPLA3 I148M and TM6SF2 E167K increase the risk of liver injury but are also associated with decreased circulating lipids and cardiovascular protection (25). This raises the possibility that therapeutic strategies targeting these genes associated with liver disease may adversely impact cardiovascular risk. However, we found that the GPAM rs10787429 allele that associated with increased liver biochemistries also associated with increased total cholesterol (P=2.0x10−17), LDL cholesterol (P=2.0x10−10), and HDL cholesterol (P=6.6x10−37) (Supplementary Table 4).
Single-cell RNA sequencing
Single-cell RNA sequencing from 28 healthy human livers (24) demonstrated hepatocyte-predominant expression of GPAM (Figure 6). An expression-based analysis revealed that GPAM was present in 48.5% of VLDL-producing cells and only 9.5% of VLDL-non-producing cells (P=9.4x10−103) (Figure 6).
Figure 6:
Single-cell RNA sequencing reveals that GPAM is predominately expressed in hepatocytes and co-expresses with VLDL production-related genes. (A) GPAM expression in parenchymal and non-parenchymal liver cells shown in purple, (B) violin plot of GPAM expression by cell identity, (C) VLDL-producing hepatocytes are defined by co-expression of more than three transcripts of APOB and MTTP, (D) GPAM expression in VLDL-producing compared to non-VLDL producing hepatocytes. GPAM was present in 48.5% of VLDL-producing cells and only 9.5% of VLDL non-producing cells (P=9.4x10−103).
DISCUSSION
In this study, we performed GWAS on circulating levels of ALT, AST, ALP and total bilirubin in UK Biobank, a large population-based cohort. Our analysis of shared GWAS risk loci for ALT and AST, as well as the results of a recent trans-ethnic study in UK Biobank (15), confirm the key role in liver injury played by PNPLA3 (11, 26), TM6SF2 (27, 28), SERPINA1 (29), HSD17B13 (13), SH2B3 (30), PANX1 (7, 31), EFNA1 (4, 32), ERLIN1 (33), AKNA (7), MTTP (34), ZNF827 (4, 7), and EFHD1 (4, 7). Consistent with the hypothesis that loci associated with ALT or AST could be related to kidney or muscle damage, we identified ANO5 rs7481951, for which homozygosity has been shown to lead to muscular dystrophy (35). Of the 18 shared genome-wide significant loci from our ALP and total bilirubin GWAS, we confirm several loci previously associated with liver injury at TM6SF2 (27, 28), HNF1A (36), ADH4 (37, 38), PCCB (7, 39), CPS1 (7, 40), and APOE (12, 41).
We focused on variants associated with elevations across all four liver biochemistries and that also associated with liver injury in Mass General Brigham Biobank, a hospital-based cohort. This identified a novel genetic locus at the mitochondrial glycerol-3-phosphate acyltransferase (GPAM rs10787429). In addition to demonstrating allele dose-dependent effects across four biomarkers of liver injury, GPAM rs10787429 also associated with several categories of liver disease based on ICD-10 diagnostic codes, demonstrated an interaction with environmental risk factors for liver disease, and associated with elevated circulating lipids. Our study is the first to perform an analysis across all four biomarkers of liver injury, and complements recent reports that have associated variation in GPAM with quantified liver fat by abdominal MRI imaging in UK Biobank (12, 42), and with severity of histologic assessment of liver steatosis in European cohorts (12).
The genetic risk locus at GPAM shares several features in common with other variants previously linked to liver disease. First, genetic variants in PNPLA3, TM6SF2, and HSD17B13 are strong genetic determinants of both fatty liver disease and alcohol-related liver cirrhosis (11, 13, 27, 28, 43). The overlap between the genetic determinants of these two diseases suggests that fatty liver, regardless of cause, is harmful and may progress to inflammation and fibrosis. Consistent with these results, we find that genetic variation in GPAM is associated with both fatty liver disease and severe alcohol-related liver cirrhosis in UK Biobank. Second, a unique feature of genetic variants associated with liver disease is a strong interaction with the environment. For example, a recent study demonstrated that adiposity amplifies the risk of fatty liver disease conferred by multiple loci: PNPLA3 I148M, TM6SF2 E167K and GCKR P446L (44). Similar to these results, we found that BMI as well as weekly alcohol use amplified the risk of hepatocellular injury associated with sequence variation at GPAM rs10787429. We suggest that certain genetic variants, in combination with an environmental trigger, such as high BMI or excess alcohol intake, act synergistically to uncover liver disease phenotypes.
The PNPLA3 I148M and TM6SF2 E167K alleles that increase the risk of cirrhosis have been reported to decrease circulating atherogenic lipid particles and are associated with lower risk of cardiovascular disease (25). This raises the possibility that therapeutic strategies targeting PNPLA3 or TM6SF2 may adversely impact cardiovascular risk. Interestingly, GPAM rs10787429 was associated with both increased risk of liver injury as well as elevated total cholesterol, LDL cholesterol, and HDL cholesterol. It is therefore plausible that therapeutic targeting of GPAM may protect against liver disease without adversely impacting the risk of cardiovascular disease, although further laboratory-based validation of this concept is required.
Glycerol-3-phosphate acyltransferase catalyzes the rate-limiting step in the de novo pathway of glycerolipid synthesis (45). Four isoforms of this enzyme have been identified; the mitochondrial glycerol-3-phosphate acyltransferase, encoded by a nuclear gene, is located in the outer mitochondrial membrane and is responsible for 30-50% of the total glycerol-3-phosphate acyltransferase activity in the liver (45). Consistent with these results, we demonstrate hepatocyte-predominant expression of GPAM in subclusters that overlap with cells involved in VLDL production. In addition to catalyzing the committed step in triacylglycerol synthesis, GPAM also directs the flux of fatty acids towards glycerolipid synthesis and away from β-oxidation (46). AMP-activated kinase, a sensor of cellular energy stores, has been shown to inhibit GPAM, thereby decreasing triacylglycerol synthesis and up-regulating β-oxidation when cellular energy stores are low (46).
Prior studies have linked GPAM to liver triglyceride content, body weight, and plasma lipid levels in murine overexpression and knockout experiments (46-48). Several small molecule inhibitors of glycerol-3-phosphate acyltransferase have been reported, including compounds with lower half maximal inhibitory concentration to GPAM (49). In diet-induced obese mice, pharmacologic inhibition of glycerol-3-phosphate acyltransferase reduced food intake, decreased body weight and adiposity, enhanced fatty acid oxidation, enhanced insulin sensitivity, and reversed hepatic steatosis (50). Together, these observations support further investigation of the role of GPAM as a therapeutic target in liver disease.
Our results should be interpreted in the context of several important limitations. Further research will be needed to confirm these results across multiple ethnicities. Among liver disease cases analyzed in this study, the presence of hepatitis B or hepatitis C was not systematically assessed. ICD-10 diagnostic codes are known to be imprecise in the context of clinical care; additional studies on the impact of the GPAM rs10787429 locus and other loci on biopsy-confirmed liver disease are warranted. We did not confirm associations with binary liver disease phenotypes or gene environment interactions in Mass General Brigham Biobank; lack of associations could be due to false positives or insufficient power, and validation in additional cohorts is warranted. Finally, there are multiple single nucleotide polymorphisms at the GPAM locus in high linkage disequilibrium; further studies will be necessary to uncover the causative variant and its functional significance.
In conclusion, we identified a common genetic variant in GPAM that is associated with susceptibility to liver injury and also associated with increased circulating lipid particles. The risk of liver injury may increase synergistically in the setting of higher BMI and increasing alcohol use. Our findings provide human genetic support for the role of GPAM in liver injury; therapeutic targeting of this enzyme should be explored as a potential approach for the treatment of liver disease.
Supplementary Material
ACKNOWLEDGMENTS AND FINANCIAL SUPPORT
This research has been conducted using the UK Biobank Resource under Application Number 20915. We thank all the UK Biobank participants whose contribution to this study lead to advancing our understanding of liver disease. Ilana Usiskin is acknowledged for R expertise. A.H. is supported by the Beth Israel Deaconess Medical Center Weener Family Residency Research Small Grants Program. M.M. is supported by NIH T32 HL007427. J.B. is supported by NIH T32 GM136651-01. J.A.L.S. is supported by R01 HL123915 and R01 HL141826. E.K.S is supported by P01 HL114501, R01 HL133135, R01 HL137927, and R01 HL147148. S.V. is supported by NIH K08 DK113109 and the Doris Duke Charitable Foundation (Grant# 2019081). Z.G.J. is supported by NIH K08 DK115883, the Alan Hofmann Clinical and Translational Research Award from American Association for the Study of Liver Diseases, and the Clinical Research Award from the American College of Gastroenterology. B.D.H. is supported by NIH K08 HL136928, U01 HL089856, R01 HL147148, and R01 HL135142. M.H.C is supported by R01 HL135142, R01 HL137927, R01 HL089856, R01 HL147148.
The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The funding body has no role in the design of the study and collection, analysis, and interpretation of data and in writing the manuscript.
Footnotes
Conflicts of interest: A.H. serves on the scientific advisory board of Olix Pharmaceuticals and consults for Deep Track Capital. E.K.S. has received grant support from GSK and Bayer. Z.G.J. consults for Olix Pharmaceuticals and Boehringer Ingelheim and receives grants from Gilead Sciences. N.H.A consults for and is on the advisory boards of TRIo healthcare, Gilead Sciences, EchoSens, Ligand Pharmaceuticals, Shionogi, Atea Pharmaceuticals, Axcella Health, GSK and Jannsen Pharmaceuticals; he is a stockholder of F-Star Therapeutics and is a Director of the Liver Institute for Education and Research. M.H.C. has received grant support from GSK and Bayer, and consulting or speaking fees from Illumina, Genentech and AstraZeneca. The remaining authors have no conflicts of interest to declare.
REFERENCES
- 1.Murphy SL, Xu J, Kochanek KD, Curtin SC, Arias E Deaths: Final Data for 2015. Natl Vital Stat Rep 2017;66:1–75. [PubMed] [Google Scholar]
- 2.Loomba R, Schork N, Chen CH, Bettencourt R, Bhatt A, Ang B, Nguyen P, et al. Heritability of Hepatic Fibrosis and Steatosis Based on a Prospective Twin Study. Gastroenterology 2015;149:1784–1793. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Nelson MR, Tipney H, Painter JL, Shen J, Nicoletti P, Shen Y, Floratos A, et al. The support of human genetic evidence for approved drug indications. Nature Genetics 2015;47:856–860. [DOI] [PubMed] [Google Scholar]
- 4.Chambers JC, Zhang W, Sehmi J, Li X, Wass MN, Van der Harst P, Holm H, et al. Genome-wide association study identifies loci influencing concentrations of liver enzymes in plasma. Nat Genet 2011;43:1131–1138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Young KA, Palmer ND, Fingerlin TE, Langefeld CD, Norris JM, Wang N, Xiang AH, et al. Genome-Wide Association Study Identifies Loci for Liver Enzyme Concentrations in Mexican Americans: The GUARDIAN Consortium. Obesity (Silver Spring) 2019;27:1331–1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Moon S, Kim YJ, Han S, Hwang MY, Shin DM, Park MY, Lu Y, et al. The Korea Biobank Array: Design and Identification of Coding Variants Associated with Blood Biochemical Traits. Sci Rep 2019;9:1382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Kanai M, Akiyama M, Takahashi A, Matoba N, Momozawa Y, Ikeda M, Iwata N, et al. Genetic analysis of quantitative traits in the Japanese population links cell types to complex human diseases. Nat Genet 2018;50:390–400. [DOI] [PubMed] [Google Scholar]
- 8.Kim YJ, Go MJ, Hu C, Hong CB, Kim YK, Lee JY, Hwang JY, et al. Large-scale genome-wide association studies in East Asians identify new genetic loci influencing metabolic traits. Nat Genet 2011;43:990–995. [DOI] [PubMed] [Google Scholar]
- 9.Prins BP, Kuchenbaecker KB, Bao Y, Smart M, Zabaneh D, Fatemifar G, Luan J, et al. Genome-wide analysis of health-related biomarkers in the UK Household Longitudinal Study reveals novel associations. Sci Rep 2017;7:11008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Yuan X, Waterworth D, Perry JRB, Lim N, Song K, Chambers JC, Zhang W, et al. Population-based genome-wide association studies reveal six loci influencing plasma levels of liver enzymes. American journal of human genetics 2008;83:520–528. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Romeo S, Kozlitina J, Xing C, Pertsemlidis A, Cox D, Pennacchio LA, Boerwinkle E, et al. Genetic variation in PNPLA3 confers susceptibility to nonalcoholic fatty liver disease. Nature genetics 2008;40:1461–1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Jamialahmadi O, Mancina RM, Ciociola E, Tavaglione F, Luukkonen PK, Baselli G, Malvestiti F, et al. Exome-wide Association Study on Alanine Aminotransferase Identifies Sequence Variants in the GPAM and APOE Associated with Fatty Liver Disease. Gastroenterology 2020. [DOI] [PubMed] [Google Scholar]
- 13.Abul-Husn NS, Cheng X, Li AH, Xin Y, Schurmann C, Stevis P, Liu Y, et al. A Protein-Truncating HSD17B13 Variant and Protection from Chronic Liver Disease. New England Journal of Medicine 2018;378:1096–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lindén D, Ahnmark A, Pingitore P, Ciociola E, Ahlstedt I, Andréasson A-C, Sasidharan K, et al. Pnpla3 silencing with antisense oligonucleotides ameliorates nonalcoholic steatohepatitis and fibrosis in Pnpla3 I148M knock-in mice. Molecular metabolism 2019;22:49–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ward LD, Tu H-C, Quenneville C, Flynn-Carroll AO, Parker MM, Deaton AM, Haslett PAJ, et al. Genome-wide association study of circulating liver enzymes reveals an expanded role for manganese transporter SLC30A10 in liver health. bioRxiv 2020:2020.2005.2019.104570. [Google Scholar]
- 16.Wroblewski F The clinical significance of alterations in transaminase activities of serum and other body fluids. Adv Clin Chem 1958;Vol. 1:313–351. [PubMed] [Google Scholar]
- 17.Ingelsson E, McCarthy MI Human Genetics of Obesity and Type 2 Diabetes Mellitus: Past, Present, and Future. Circ Genom Precis Med 2018;11:e002090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bycroft C, Freeman C, Petkova D, Band G, Elliott LT, Sharp K, Motyer A, et al. The UK Biobank resource with deep phenotyping and genomic data. Nature 2018;562:203–209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Song W, Torous J, Kossowsky J, Chen C-Y, Huang H, Wright A Genome-wide association analysis of insomnia using data from Partners Biobank. Scientific Reports 2020;10:6928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience 2015;4:7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bulik-Sullivan BK, Loh PR, Finucane HK, Ripke S, Yang J, Patterson N, Daly MJ, et al. LD Score regression distinguishes confounding from polygenicity in genome-wide association studies. Nat Genet 2015;47:291–295. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Watanabe K, Taskesen E, van Bochoven A, Posthuma D Functional mapping and annotation of genetic associations with FUMA. Nature Communications 2017;8:1826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Tomson P A GREAT AND GROWING EVIL: The medical consequences of alcohol abuse. The Journal of the Royal College of General Practitioners 1987;37:469–469. [Google Scholar]
- 24.Brancale J, Vilarinho S A single cell gene expression atlas of 28 human livers. Journal of Hepatology 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Liu DJ, Peloso GM, Yu H, Butterworth AS, Wang X, Mahajan A, Saleheen D, et al. Exome-wide association study of plasma lipids in >300,000 individuals. Nature genetics 2017;49:1758–1766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.BasuRay S, Wang Y, Smagris E, Cohen JC, Hobbs HH Accumulation of PNPLA3 on lipid droplets is the basis of associated hepatic steatosis. Proc Natl Acad Sci U S A 2019;116:9521–9526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kozlitina J, Smagris E, Stender S, Nordestgaard BG, Zhou HH, Tybjærg-Hansen A, Vogt TF, et al. Exome-wide association study identifies a TM6SF2 variant that confers susceptibility to nonalcoholic fatty liver disease. Nature genetics 2014;46:352–356. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Dongiovanni P, Petta S, Maglio C, Fracanzani AL, Pipitone R, Mozzi E, Motta BM, et al. Transmembrane 6 superfamily member 2 gene variant disentangles nonalcoholic steatohepatitis from cardiovascular disease. Hepatology 2015;61:506–514. [DOI] [PubMed] [Google Scholar]
- 29.Schneider CV, Hamesch K, Gross A, Mandorfer M, Moeller LS, Pereira V, Pons M, et al. Liver Phenotypes of European Adults Heterozygous or Homozygous for Pi*Z Variant of AAT (Pi*MZ vs Pi*ZZ genotype) and Non-carriers. Gastroenterology 2020. [DOI] [PubMed] [Google Scholar]
- 30.Mori T, Suzuki-Yamazaki N, Takaki S Lnk/Sh2b3 Regulates Adipose Inflammation and Glucose Tolerance through Group 1 ILCs. Cell Rep 2018;24:1830–1841. [DOI] [PubMed] [Google Scholar]
- 31.Crespo Yanguas S, da Silva TC, Pereira IVA, Maes M, Willebrords J, Shestopalov VI, Goes BM, et al. Genetic ablation of pannexin1 counteracts liver fibrosis in a chemical, but not in a surgical mouse model. Archives of toxicology 2018;92:2607–2627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Iida H, Honda M, Kawai HF, Yamashita T, Shirota Y, Wang BC, Miao H, et al. Ephrin-A1 expression contributes to the malignant characteristics of {alpha}-fetoprotein producing hepatocellular carcinoma. Gut 2005;54:843–851. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Feitosa MF, Wojczynski MK, North KE, Zhang Q, Province MA, Carr JJ, Borecki IB The ERLIN1-CHUK-CWF19L1 gene cluster influences liver fat deposition and hepatic inflammation in the NHLBI Family Heart Study. Atherosclerosis 2013;228:175–180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Newberry EP, Xie Y, Kennedy SM, Graham MJ, Crooke RM, Jiang H, Chen A, et al. Prevention of hepatic fibrosis with liver microsomal triglyceride transfer protein deletion in liver fatty acid binding protein null mice. Hepatology 2017;65:836–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hicks D, Sarkozy A, Muelas N, Köehler K, Huebner A, Hudson G, Chinnery PF, et al. A founder mutation in Anoctamin 5 is a major cause of limb girdle muscular dystrophy. Brain 2010;134:171–182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ni Q, Ding K, Wang KQ, He J, Yin C, Shi J, Zhang X, et al. Deletion of HNF1α in hepatocytes results in fatty liver-related hepatocellular carcinoma in mice. FEBS Lett 2017;591:1947–1957. [DOI] [PubMed] [Google Scholar]
- 37.Gelernter J, Kranzler HR, Sherva R, Almasy L, Koesterer R, Smith AH, Anton R, et al. Genome-wide association study of alcohol dependence:significant findings in African- and European-Americans including novel risk loci. Mol Psychiatry 2014;19:41–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Deltour L, Foglio MH, Duester G Metabolic deficiencies in alcohol dehydrogenase Adh1, Adh3, and Adh4 null mutant mice. Overlapping roles of Adh1 and Adh4 in ethanol clearance and metabolism of retinol to retinoic acid. J Biol Chem 1999;274:16796–16801. [DOI] [PubMed] [Google Scholar]
- 39.Wongkittichote P, Ah Mew N, Chapman KA Propionyl-CoA carboxylase – A review. Molecular Genetics and Metabolism 2017;122:145–152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Park M-J, D’Alecy LG, Anderson MA, Basrur V, Feng Y, Brady GF, Kim D-i, et al. Constitutive release of CPS1 in bile and its role as a protective cytokine during acute liver injury. Proceedings of the National Academy of Sciences 2019;116:9125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Schierwagen R, Maybüchen L, Zimmer S, Hittatiya K, Bäck C, Klein S, Uschner FE, et al. Seven weeks of Western diet in apolipoprotein-E-deficient mice induce metabolic syndrome and non-alcoholic steatohepatitis with liver fibrosis. Scientific reports 2015;5:12931–12931. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Haas ME, Pirruccello JP, Friedman SN, Emdin CA, Ajmera VH, Simon TG, Homburger JR, et al. Machine learning enables new insights into clinical significance of and genetic contributions to liver fat accumulation. medRxiv 2020:2020.2009.2003.20187195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tian C, Stokowski RP, Kershenobich D, Ballinger DG, Hinds DA Variant in PNPLA3 is associated with alcoholic liver disease. Nat Genet 2010;42:21–23. [DOI] [PubMed] [Google Scholar]
- 44.Stender S, Kozlitina J, Nordestgaard BG, Tybjærg-Hansen A, Hobbs HH, Cohen JC Adiposity amplifies the genetic risk of fatty liver disease conferred by multiple loci. Nat Genet 2017;49:842–847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Coleman RA, Mashek DG Mammalian triacylglycerol metabolism: synthesis, lipolysis, and signaling. Chem Rev 2011;111:6359–6386. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Hammond LE, Gallagher PA, Wang S, Hiller S, Kluckman KD, Posey-Marcos EL, Maeda N, et al. Mitochondrial glycerol-3-phosphate acyltransferase-deficient mice have reduced weight and liver triacylglycerol content and altered glycerolipid fatty acid composition. Mol Cell Biol 2002;22:8204–8214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Neschen S, Morino K, Hammond LE, Zhang D, Liu ZX, Romanelli AJ, Cline GW, et al. Prevention of hepatic steatosis and hepatic insulin resistance in mitochondrial acyl-CoA:glycerol-sn-3-phosphate acyltransferase 1 knockout mice. Cell Metab 2005;2:55–65. [DOI] [PubMed] [Google Scholar]
- 48.Lindén D, William-Olsson L, Ahnmark A, Ekroos K, Hallberg C, Sjögren HP, Becker B, et al. Liver-directed overexpression of mitochondrial glycerol-3-phosphate acyltransferase results in hepatic steatosis, increased triacylglycerol secretion and reduced fatty acid oxidation. The FASEB Journal 2006;20:434–443. [DOI] [PubMed] [Google Scholar]
- 49.Wydysh EA, Medghalchi SM, Vadlamudi A, Townsend CA Design and synthesis of small molecule glycerol 3-phosphate acyltransferase inhibitors. J Med Chem 2009;52:3317–3327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Kuhajda FP, Aja S, Tu Y, Han WF, Medghalchi SM, El Meskini R, Landree LE, et al. Pharmacological glycerol-3-phosphate acyltransferase inhibition decreases food intake and adiposity and increases insulin sensitivity in diet-induced obesity. Am J Physiol Regul Integr Comp Physiol 2011;301:R116–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





