Skip to main content
Erschienen in: BMC Medicine 1/2023

Open Access 01.12.2023 | Research article

Identification of genetic variants associated with diabetic kidney disease in multiple Korean cohorts via a genome-wide association study mega-analysis

verfasst von: Heejin Jin, Ye An Kim, Young Lee, Seung-hyun Kwon, Ah Ra Do, Sujin Seo, Sungho Won, Je Hyun Seo

Erschienen in: BMC Medicine | Ausgabe 1/2023

Abstract

Background

The pathogenesis of diabetic kidney disease (DKD) is complex, involving metabolic and hemodynamic factors. Although DKD has been established as a heritable disorder and several genetic studies have been conducted, the identification of unique genetic variants for DKD is limited by its multiplex classification based on the phenotypes of diabetes mellitus (DM) and chronic kidney disease (CKD). Thus, we aimed to identify the genetic variants related to DKD that differentiate it from type 2 DM and CKD.

Methods

We conducted a large-scale genome-wide association study mega-analysis, combining Korean multi-cohorts using multinomial logistic regression. A total of 33,879 patients were classified into four groups—normal, DM without CKD, CKD without DM, and DKD—and were further analyzed to identify novel single-nucleotide polymorphisms (SNPs) associated with DKD. Additionally, fine-mapping analysis was conducted to investigate whether the variants of interest contribute to a trait. Conditional analyses adjusting for the effect of type 1 DM (T1D)-associated HLA variants were also performed to remove confounding factors of genetic association with T1D. Moreover, analysis of expression quantitative trait loci (eQTL) was performed using the Genotype-Tissue Expression project. Differentially expressed genes (DEGs) were analyzed using the Gene Expression Omnibus database (GSE30529). The significant eQTL DEGs were used to explore the predicted interaction networks using search tools for the retrieval of interacting genes and proteins.

Results

We identified three novel SNPs [rs3128852 (P = 8.21×10−25), rs117744700 (P = 8.28×10−10), and rs28366355 (P = 2.04×10−8)] associated with DKD. Moreover, the fine-mapping study validated the causal relationship between rs3128852 and DKD. rs3128852 is an eQTL for TRIM27 in whole blood tissues and HLA-A in adipose-subcutaneous tissues. rs28366355 is an eQTL for HLA-group genes present in most tissues.

Conclusions

We successfully identified SNPs (rs3128852, rs117744700, and rs28366355) associated with DKD and verified the causal association between rs3128852 and DKD. According to the in silico analysis, TRIM27 and HLA-A can define DKD pathophysiology and are associated with immune response and autophagy. However, further research is necessary to understand the mechanism of immunity and autophagy in the pathophysiology of DKD and to prevent and treat DKD.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12916-022-02723-4.
Heejin Jin and Ye An Kim contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
CKD
Chronic kidney disease
DEGs
Differentially expressed genes
DKD
Diabetic kidney disease
DM
Diabetes mellitus
eGFR
Estimated glomerular filtration rate
eQTL
Expression quantitative trait loci
ESRD
End-stage renal disease
GEO
Gene Expression Omnibus
GTEx
Genotype-Tissue Expression
GWAS
Genome-wide association study
HbA1c
Glycosylated hemoglobin
HWE
Hardy–Weinberg equilibrium
IgAN
IgA nephropathy
KDIGO
Kidney Disease: Improving Global Outcomes
KEGG
Kyoto Encyclopedia of Genes and Genomes
KoGES
Korean Genome and Epidemiology Study
LD
Linkage disequilibrium
MDS
Multidimensional scaling
MLR
Multinomial logistic regression
SD
Standard deviation
SNP
Single-nucleotide polymorphism
STRING
Search Tool for the Retrieval of Interacting Genes/Proteins
VHSMC
Veterans Health Service Medical Center
VIF
Variance inflation factor

Background

Diabetic kidney disease (DKD) is the primary etiology of chronic kidney disease (CKD) in patients with diabetes mellitus (DM) [1] and the leading cause of CKD and end-stage renal disease (ESRD) in most developed countries [2]. Moreover, risk factors for the development of DKD in patients with type 2 DM include cardiovascular risk factors such as high urinary albumin creatinine ratio, old age, hyperglycemia, and hypertension [3]. Although the importance of hyperglycemia in the development of DKD has been illustrated in several studies [46], some patients with type 2 DM experience a relatively rapid deterioration in renal function, whereas others maintain normal renal function even with suboptimal glycemic levels [7]. Furthermore, patients with DKD showed familial clustering [8] and ethnic group differences [9, 10]. This susceptibility highlights the need to identify the specific genetic factors that affect the onset and progression of DKD in patients with DM.
Recently, genome-wide association studies (GWASs) have identified more than 33 genes for DKD in type 2 DM, including APOL1, GABRR1, GCKR, and UMOD [1119]. Moreover, most of the genes reportedly associated with DKD need to be confirmed by further replication studies and a detailed analysis of their functional role in DKD using experimental models [20]. Two complex fundamental features define DKD: the decline of estimated glomerular filtration rate (eGFR) and the presence of proteinuria; hence, it would be better to combine these phenotypes during analysis, to incorporate DM and hypertension-related CKD. However, previous research on GWAS phenotypes for DKD or CKD was confined to the assessment of single phenotypes, such as uric acid, eGFR, ESRD, and proteinuria, and failed to focus on defining DKD [1119, 21]. Hypertension is both an underlying risk factor and a consequence of DKD due to persistent high blood pressure in the arteries around the kidney [22]. Additionally, up to 75% of patients with DM also experience hypertension, and individuals with only hypertension frequently exhibit signs of insulin resistance [23, 24]. Although previous studies were singularly focused on either CKD or DKD, genes such as UMOD were linked to both hypertensive CKD (non-DKD) and DKD [20, 25, 26]. Hence, we hypothesized that single-nucleotide polymorphism (SNP)-related traits for DKD could be discovered through a GWAS mega-analysis using multinomial logistic regression (MLR).
In addition, although there are few studies on decreased renal function in middle Eastern descent [21], Japanese [17, 27] and Han Chinese [18] populations, most studies were conducted on the European and African-American populations [7, 1216, 19]. There is a need for GWAS on DKD in large-scale Korean multi-cohorts. As reported in previous studies by the Veterans Health Service Medical Center (VHSMC) [28, 29], several elderly veterans are diagnosed with DKD due to the extended duration of type 2 DM. Consequently, studies on DKD in Korean multi-cohorts would be helpful. Toward this goal, we conducted a large-scale GWAS mega-analysis of multi-cohorts, combining the VHSMC cohorts and Korean Genome and Epidemiology Study (KoGES) consortium using MLR with four groups: normal, DM without CKD (“only DM”), CKD without DM (“only CKD”) and DKD.

Methods

Study population

Clinical and genetic data from multi-cohorts of the VHSMC cohort and the KoGES consortium were integrated in this study (sample size, n = 81,039, Fig. 1). In the previously constructed VHSMC cohort, diagnosed with type 2 DM by VHSMC endocrinologists [28, 29], those who met the inclusion criteria (n = 916) were enrolled in this study. The KoGES consortium is a nationwide cohort representative of genome research in Korea [30], of which three cohorts related to population-based studies [Korean Association Resource from Ansan and Ansung (KARE, n = 8840) cohort, KoGES Health Examinees (HEXA, n = 61,568) cohort, and KoGES cardiovascular disease association study (CAVAS, n = 9,715) cohort] were enrolled in this study. This study excluded subjects without Korea Biobank Array genotype data or phenotype (DM, eGFR, albuminuria, and hypertension) data, those who had chronic diseases affecting DM (kidney cancer, pancreatic disease, etc.) and renal function (liver cancer, chemotherapy, etc.), and those younger than 65 years for the control group from the VHSMC cohort and KoGES consortium. After applying the exclusion criteria, 30,069 participants were included in the GWAS mega-analysis (Fig. 1). The VHSMC cohort contained patients diagnosed with type 2 DM and hypertension by certified doctors, whereas in the KoGES cohort data, DM was defined by any of the following four categories (Table 1): (1) DM diagnosis checked in the questionnaire, (2) blood glucose levels ≥ 200 mg/dL 2 h after glucose loading, (3) glycosylated hemoglobin (HbA1c) amount ≥ 6.5%, and (4) overnight fasting blood glucose levels ≥ 126 mg/dL. To develop a distinct control group, participants aged 65 years or above who did not have DM or renal failure were included in the analysis.
Table 1
Criteria for the outcome groups for multinomial logistic regression
 
Satisfied with any of the following?
    
1
1) Checking in DM diagnosis of the questionnaire
2) 2 h loading of blood glucose levels ≥ 200 mg/dL
3) HbA1c ≥ 6.5%
4) Overnight fasting blood glucose levels ≥ 126 mg/dL
  
2
1) Creatinine 1.3 mg/dL (men), 1.0 mg/dL (women)
2) eGFR < 60 mL/min/1.73 m2
3) Albuminuria
 
 
Group
DKD (1)
Only DM (2)
Only CKD (3)
Control (4)
HbA1C Glycosylated hemoglobin

DKD phenotype definition

The eGFR was calculated using the abbreviated Modification of Diet in Renal Disease equation as follows: 175 × serum creatinine−1.154 × age−0.203 (×0.742 if female) [31]. According to the Kidney Disease: Improving Global Outcomes (KDIGO) guidelines, eGFR and albuminuria categories were used to assess a renal complication [32]. Albuminuria was classified into three categories based on the following albumin-to-creatinine ratios: < 3 mg/mmol creatinine, normal to mildly increased; 3–29 mg/mmol, moderately increased; and ≥ 30 mg/mmol creatinine, severely increased. Since the dysfunction of the glomerular barrier (represented by proteinuria) and reduced renal function (assessed using the eGFR) may develop independently, the various phenotypes for DKD were defined as follows (Table 1): (1) creatinine level > 1.3 mg/dL (men), > 1.0 mg/dL (women), (2) eGFR < 60 mL/min/1.73 m2, and (3) albuminuria.

Ethics

The institutional review board (IRB) at the Veterans Health Service Medical Center approved the study protocols for the VHSMC cohorts after obtaining informed consent (IRB No. 2018-08-032 and IRB No. 2020-02-031). However, the IRB approved the protocol for the KoGES consortium (IRB No. 2021-05-007) after waiving the need for informed consent since this was a retrospective study; the committee of the National Biobank of Korea, the Korea Disease Control and Prevention Agency (KBN-2021-042), approved the use of bioresources in this study. This study was conducted in compliance with the Declaration of Helsinki.

Genotyping and imputation

Genomic DNA was extracted from venous blood samples, and 100 ng DNA was genotyped using the c Affymetrix Axiom 1.1 (Affymetrix, Santa Clara, CA) [33]. The genotypes were identified using a K-medoid clustering-based algorithm to minimize the batch effect [34]. The PLINK (version 1.9, Boston, MA) and ONETOOL [35] software packages were used for quality control processes. We excluded samples matching any of the following criteria: (1) sex inconsistencies or (2) a call rate of up to 95%. Furthermore, SNPs were filtered out if (1) the call rate was lower than 95% or (2) the Hardy–Weinberg equilibrium (HWE) test showed P < 1×10−5. The genotype imputation was conducted using the Northeast Asian Reference Database imputation server (https://​nard.​macrogen.​com/​), and data of 1779 Northeast Asians [36] were used for the reference panel. Pre-phasing and imputation were performed using Eagle v2.4 [37] and Minimac4 [38], respectively. Post imputation, imputed SNPs were removed if the R-squared value was less than 0.8, there were duplicated SNPs, missing genotype rates were more than 0.05, P-values for HWE were less than 1×10−5, or minor allele frequencies were less than 0.01. After quality control and imputation, 6,159,267 SNPs were selected for association analyses.

Genome-wide association analysis

Baseline characteristics of the study population have been reported as means ± standard deviation (SD) for continuous variables and numbers and as proportions for categorical variables. Genome-wide association analyses were conducted with an MLR model for the categorical response variable with four levels (normal, only DM, only CKD, and DKD), implemented in SNPTEST v2.5.6 [39]. We evaluated the overall fit of the model by comparing the likelihood of the two models: a full model with genotype risk factors and a reduced model with covariates only. Age, sex, and ten principal component scores were selected as covariates. To estimate the marginal genetic effects for six contrasts [contrast (1) only DM vs. normal, (2) only CKD vs. normal, (3) only DKD vs. normal, (4) only CKD vs. only DM, (5) DKD vs. only DM, and (6) DKD vs. only CKD], a Wald test was used. To verify that there was no confounding due to population stratification in this study, the variance inflation factor (VIF) was calculated, whereby a VIF value close to 1 indicated no genomic inflation. The regional plot for significant genetic variation was generated using the LocusZoom software with linkage disequilibrium (LD) information of East Asians from the 1000 Genomes Project [40]. The bottom panel displays gene symbols and the location within the region, derived from 1000 genomes (ASN hg19/Nov2014). The threshold for statistical significance in this model was P < 5.0×10−8, which is conventionally considered to reflect genome-wide significance. To estimate the relative proportion of phenotypic variance explained by all observed common SNPs, genome-wide complex trait analysis (GCTA v1.91.7) was used for heritability calculation [41].

Conditional analyses

HLA has been reported to have an effect on T1D, and variants in the HLA region were adjusted to remove the confounding effects of T1D on DKD. A conditional MLR model was applied, adjusting the effect of T1D-associated HLA variants. Among 35 previously reported T1D-associated HLA variants, one SNP (rs9275490) in DR-DQ loci and one SNP (rs9271346) in non-DR-DQ loci were included as covariates for the analysis [42]. Furthermore, to determine the variants that affect DKD independently of DM and CKD, we performed additional logistic regression analyses for DKD after adjusting for the effects of DM and CKD.

Fine-mapping analysis

To prioritize whether the variants discovered are candidate causal variants, fine-mapping analysis was performed. Among significant GWAS hits, even after adjusting for effects of T1D-associated HLA variants, DM, and CKD, only SNPs with genome-wide significantly related to DKD were used for fine-mapping analysis. For each target variant, we first selected the set of SNPs, consisting of the most significant SNP and a 100-kb window of SNPs around it. The LD matrix between SNPs was computed using PLINK (version 1.9) [43]. Using a Bayesian approach (PAINTOR method) [44], we estimated the posterior probabilities of causative SNPs at a given fine-mapping locus.

Functional annotation analyses

The eQTL analysis was performed using the Genotype-Tissue Expression (GTEx) dataset. To identify significant eQTL genes, it was assumed that approximately 200,000 SNPs were used in the eQTL analysis considering 20,000 genes and LD block [45]. Therefore, using the Bonferroni correction, we set the significance threshold to 0.05/200,000 (=2.5×10−7). We used the LocusFocus tool to generate a colocalization plot, showing the lead SNP responsible for both GWAS and eQTL signals at loci [40]. GTEx version V7 and 1000 Genomes Phase 3 East Asian LD were used for this plot. The associated genes were further investigated for differently expressed genes (DEGs) in the glomeruli of patients with DM while controlling for age from the Gene Expression Omnibus (GEO) dataset (GSE30529). The platform used for the analysis of GSE30529 was the GPL571 [HG-U133A_2] Affymetrix Human Genome U133A 2.0 Array, which included genes from the kidneys of ten subjects with diabetes and twelve control samples of genes from the kidneys of healthy people. Furthermore, the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) open-access database was used to identify biological functions based on the identified genes [46]. The minimum required interaction score was set as 0.4 (medium confidence). The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis [47] was also conducted using the STRING database.

Transcriptome-wide association analysis

Gene-based association analyses were performed for 7254 protein-coding genes using PrediXcan [48]. We imputed tissue-specific (whole blood) gene expression variation from GWAS summary statistics using weights derived from a reference transcriptome dataset provided by PrediXcan. Statistical analyses were performed with the “limma” package in R (http://​www.​bioconductor.​org/​) [49]. Transcriptome-wide significance was defined at P = 6.89×10−6 (0.05/7254) via the Bonferroni correction.

Results

Clinical characteristics of the study participants

A total of 33,879 subjects were enrolled in this study, and the baseline characteristics of the study population are presented in Table 2. Among them, 15,664 subjects (46%) were part of the control group, whereas 8844 (26%), 6839 (20%), 2532 (8%) were part of the only DM, only CKD, and DKD groups, respectively. The mean age of the multi-cohort was 66.19 years; the VHSMC cohort had the highest mean age of 73.93 years, followed by CAVAS cohort with the mean age of 68.81 years, KARE cohort with the mean age of 68.50 years, and HEXA cohort with the mean age of 64.57 years. The proportion of male subjects was 45% of the multi-cohort; most of the VHSMC cohort were male (88%), whereas the KARE, HEXA, and CAVAS cohorts had 46, 56, and 42% male subjects, respectively. The mean body mass index (BMI) for the multi-cohort was 24.47, whereas the mean BMI for each cohort showed a similar distribution—25.30, VHSMC; 24.85, KARE; 24.33, HEXA; and 24.57, CAVAS. The proportion of DM subjects in the multi-cohort was 34%, whereas in the individual cohorts, it was 86, 39, 33, and 24% in VHSMC, KARE, HEXA, and CAVAS, respectively. In addition, hypertension showed a similar trend as diabetes. The proportion of patients with hypertension in the multi-cohort was 32%, whereas in the individual cohorts, it was 83, 37, 28, and 32% in VHSMC, KARE, HEXA, and CAVAS. The mean HbA1c (%) for the multi-cohort was 6.09. The VHSMC cohort with the highest proportion of patients with DM had the highest mean HbA1c (%) at 7.63. The mean HbA1c (%) for the KARE, HEXA, and CAVAS cohorts was 6.22, 6.05, and 5.94, respectively. The mean creatinine (mg/dL) level for the multi-cohort was 0.93, with the highest mean creatinine level detected in the VHSMC cohort at 1.61. The rest of the cohorts showed a similar trend for mean creatinine (mg/dL): KARE, 1.01; HEXA, 0.86; and CAVAS, 1.00. Furthermore, the mean eGFR (mL/min/1.73 m2) level for the multi-cohort was 76.18. However, this biomarker varied significantly based on the individual cohorts: VHSMC, 56.12; KARE, 67.25; HEXA, 82.10; and CAVAS, 65.78. The average uric acid level (mg/dL) was 4.98 for the multi-cohort, whereas for the individual cohorts, it was 5.74, VHSMC; 5.10, KARE; 4.92, HEXA; and 5.01, CAVAS. Although the number of patients with proteinuria was 7% overall and 4% in KARE, 7% in HEXA, and 5% in CAVAS cohorts, it was significantly higher in the VHSMC cohort at 44%.
Table 2
Baseline characteristics of the study populations (total sample size = 33,879)
 
Entire
VHSMC cohort
KoGES Consortium
P
KARE cohort
HEXA cohort
CAVAS cohort
Sample size
33,879
908
4,899
21,693
6,379
-
Age (years)
66.19 ± 8.00
73.93 ± 5.93
68.50 ± 8.40
64.57 ± 7.54
68.81 ± 7.67
<0.001a
Sex (male %)
15,332 (45%)
800 (88%)
2271 (46%)
12,117 (56%)
2685 (42%)
<0.001b
BMI (kg/m2)
24.47 ± 3.06
25.30 ± 2.89
24.85 ± 3.19
24.33 ± 3.02
24.57 ± 3.12
<0.001a
DM (%)
11,376 (34%)
777 (86%)
1916 (39%)
7158 (33%)
1525 (24%)
<0.001b
Hypertension (%)
10,715 (32%)
752 (83%)
1803 (37%)
6,116 (28%)
2044 (32%)
<0.001b
HbA1c (%)
6.09 ± 1.04
7.63 ± 1.74
6.22 ± 1.17
6.05 ± 1.03
5.94 ± 0.85
<0.001a
Creatinine (mg/dL)
0.93 ± 0.42
1.61 ± 1.60
1.01 ± 0.47
0.86 ± 0.39
1.00 ± 0.34
<0.001a
eGFR (mL/min/1.73 m2)
76.18 ± 16.80
56.12 ± 22.47
67.25 ± 15.12
82.10 ± 18.24
65.78 ± 12.40
<0.001a
Uric acid (mg/dL)
4.98 ± 1.38
5.74 ± 1.55
5.10 ± 1.45
4.92 ± 1.34
5.01 ± 1.43
<0.001a
Proteinuria
     
<0.001b
 No
29,690 (88%)
491 (54%)
4651 (95%)
19,726 (91%)
4822 (76%)
-
 Yes
2425 (7%)
403 (44%)
185 (4%)
1549 (7%)
288 (5%)
-
MLR group (%)
     
<0.001b
 Normal
15,664 (46%)
72 (8%)
1968 (40%)
10,518 (49%)
3106 (49%)
-
 Only DM
8844 (26%)
330 (36%)
1459 (30%)
5978 (28%)
1077 (17%)
-
 Only CKD
6839 (20%)
59 (7%)
1015 (21%)
4017 (18%)
1748 (27%)
-
 DKD
2532 (8%)
447 (49%)
457 (9%)
1180 (5%)
448 (7%)
-
VHSMC Veterans Health Service Medical Center, KoGES Korean Genome and Epidemiology Study, KARE KoGES Ansan and Ansung, HEXA KoGES Health Examinees, CAVAS KoGES cardiovascular disease association study
aANOVA test, bChi-square test

Genome-wide association analysis for DKD

We conducted a GWAS mega-analysis using MLR on 6,159,267 SNPs from 30,069 subjects from the VHSMC cohort and the KoGES consortium, and they were analyzed after the genotype quality control (Fig. 1). First, we investigated genetic variants that significantly differed between the four groups, using the likelihood ratio test (LRT). Three genetic variants passed the genome-wide significant threshold (P <5.00×10−8) and were identified as novel variations for DKD. The most significant SNP was rs3128852 near OR5V1 (LRT P = 8.21×10−25), followed by rs117744700 near HIATL1 (LRT P = 8.28×10−10), and rs28366355 near human leukocyte antigens HLA-DRB1 and HLA-DQA1 (LRT P = 2.04×10−8; (Table 3 and Additional file 1: Table S1). Notably, rs3128852 and rs117744700 were significant in DKD even after adjusting for the association of the T1D-associated HLA variants, whereas rs28366355 in the HLA region lost significant association (Additional file 1: Table S2). Furthermore, we confirmed that rs3128852 and rs117744700 were associated with DKD independently of the link between DKD and DM or CKD (Additional file 1: Table S3). Figure 2 represents the quantile–quantile (Q-Q) plot, which verified that there was no inflation in the test statistics (VIF=1.025), and Manhattan plots of the results of the MLR GWAS mega-analysis. Regional association plots are shown in Fig. 3. In addition, two more suggestive variants (P <1.00×10−6) were identified: rs1824125 near PGR (LRT P = 6.58×10−7) and rs75292524 near PARD3B and NRP2 (LRT P = 7.82×10−7; Table 3). The Q-Q and Manhattan plots for six contrasts for each pair are shown in Additional file 2: Fig. S1. Since the HLA region has a notably higher level of variability than the rest of the genome, it was confirmed through the multidimensional scaling (MDS) plot that the false-positive association was not caused by population stratification (Additional file 2: Fig. S2). Furthermore, fine-mapping results supported that the top SNP (rs3128852) with the highest posterior probability (almost 1.0) is likely to have a potential causal effect on DKD (Additional file 1: Table S4 and Additional file 2: Fig. S3A). However, the posterior probability of the second (rs117744700) significant variants was almost zero (Additional file 2: Fig. S3B). These SNPs may not actually be potential causal variants, or the causality may not be estimated owing to the absence of SNPs with high LD relationships around the target SNP (Fig. 3B, C). Since GWAS is designed for identifying variants associated with the phenotype of interest, and not causal variants, the top SNP can be interpreted as a potential causal SNP for DKD and a related SNP for the other two SNPs. The eQTL colocalization plots for potential causal variants of DKD are shown in Additional file 2: Fig. S4.
Table 3
Results of the GWAS mega-analysis using multinomial logistic regression (leading SNPs, top five)
Chr
Position
rsID
Allelea
AF
GWAS
Gene symbol
 
OR (95% CI)
P
6
29364135
rs3128852
C/T
0.281
  
8.21×1025
OR5V1 (intronic)
- only DM vs. normal
1.079 (1.030–1.132)
5.81 × 10−4
 
- only CKD vs. normal
0.977 (0.934–1.024)
0.342
 
- DKD vs. normal
1.421 (1.296–1.572)
2.32 × 10−24
 
- only CKD vs. only DM
0.905 (0.859–0.955)
2.28 ×10−4
 
- DKD vs. only DM
1.317 (1.226–1.416)
4.72 × 10−14
 
- DKD vs. only CKD
1.455 (1.351–1.567)
3.13 ×10−23
 
9
97149910
rs117744700
G/C
0.249
  
8.28×1010
HIATL1 (intronic)
- only DM vs. normal
0.950 (0.911–0.993)
0.030
 
- only CKD vs. normal
0.967 (0.926–1.017)
0.216
 
- DKD vs. normal
0.769 (0.725–0.818)
6.22 ×10−11
 
- only CKD vs. only DM
1.020 (0.966–1.078)
0.481
 
- DKD vs. only DM
0.809 (0.745–0.879)
5.22 ×10−7
 
- DKD vs. only CKD
0.793 (0.729–0.863)
7.65 ×10−8
 
6
32565056
rs28366355
G/T
0.498
  
2.04×108
HLA-DRB1(dist=7443),
- only DM vs. normal
1.117 (1.070–1.169)
3.38×108
HLA-DQA1(dist=40127)
- only CKD vs. normal
1.085 (1.037–1.137)
1.46×104
(intergenic)
- DKD vs. normal
1.114 (1.040–1.201)
9.52×104
 
- only CKD vs. only DM
0.971 (0.925–1.018)
0.224
 
- DKD vs. only DM
0.998 (0.931–1.068)
0.941
 
- DKD vs. only CKD
1.027 (0.958–1.102)
0.448
 
11
100902053
rs1824125
A/C
0.248
  
6.58 × 10−7
PGR (UTR3)
- only DM vs. normal
1.075 (1.026–1.131)
1.50 × 10−3
 
- only CKD vs. normal
1.030 (0.981–1.084)
0.229
 
- DKD vs. normal
1.211 (1.114–1.327)
1.59 × 10−7
 
- only CKD vs. only DM
0.957 (0.907–1.012)
0.118
 
- DKD vs. only DM
1.126 (1.043–1.215)
2.14×10−3
 
- DKD vs. only CKD
1.176 (1.088–1.272)
4.58×10−5
 
2
206490310
rs75292524
A/G
0.283
  
7.82×10−7
PARD3B(dist=5424),
- only DM vs. normal
0.960 (0.921–1.002)
0.072
NRP2(dist=56914)
- only CKD vs. normal
0.991 (0.947–1.039)
0.720
(intergenic)
- DKD vs. normal
0.815 (0.768–0.867)
7.82×10−7
 
- only CKD vs. only DM
1.126 (1.043–1.215)
0.239
 
- DKD vs. only DM
0.848 (0.784–0.918)
4.06×10−5
 
- DKD vs. only CKD
0.822 (0.759–0.891)
1.59×10−6
 
AF Allele frequency, CKD Chronic kidney disease, Dist Distance, DM Diabetes mellitus, DKD Diabetic kidney disease, SNP Single-nucleotide polymorphism, OR Odds ratio
aMajor/minor allele

Functional annotation

The most significant variant (rs3128852) was an eQTL for TRIM27 (tripartite motif-containing 27) in whole blood (P = 2.10×10−9) and adipose-subcutaneous cells (P = 1.60×10−7; Table 4). However, the second significant variant (rs117744700) did not have any eQTL-associated genes in the GTEx database. The third significant variant (rs28366355) was an eQTL for CYP21A1P, HLA-DQA1, HLA-DQA2, HLA-DQB1, HLA-DQB2, HLA-DRB1, HLA-DRB6, and XXbac-BPG154L12.4 in most tissues. The eQTL analysis results for major tissues that may be associated with renal function (whole blood; artery-aorta, artery-tibial; adipose-subcutaneous; heart-left ventricle; renal-cortex, adrenal gland) are presented in Table 4 and Additional file 1: Table S5. Colocalization plots for the top SNP (rs3128852) in the TRIM27 and HLA-A gene and a third significant SNP (rs28366355) in the HLA region (from 29,677,984 to 33,485,635 bp in chromosome 6) are shown in Additional file 2: Fig. S4. Moreover, the decreased expression levels of TRIM27 [estimate of the log2-fold-change (logFC) corresponding to the DKD control glomeruli was −0.506 and P = 4.10×10−3], HLA-A (logFC = −1.053 and P = 1.19×10−3), HLA-DQA1 (logFC =−1.758 and P = 8.09×10−3), HLA-DQB1 (logFC = −1.234 and P = 4.78×10−4), HLA-DQB2 (logFC = −0.450 and P = 2.21×10−2), and HLA-DRB6 (logFC =−0.725 and P = 3.53×10−5) genes were associated with DKD glomeruli in GEO datasets (GSE30529; Table 5 and Additional file 1: Table S6). Except for HLA-DRB6, which cannot be annotated in the STRING database, five genes were used as input genes in the STRING database to identify known and predicted biological functional networks. The HLA genes (HLA-A, HLA-DQA1, HLA-DQB1, and HLA-DQB2) had a strong interaction with each other (interaction score > 0.75), but TRIM27 did not interact with any of these genes (Additional file 2: Fig. S5). Although 10 KEGG pathways were associated with this network, only two genes (HLA-DQA1 and HLA-A) were annotated for DKD, which are associated with the immune response related to DKD pathogenesis (Table 6). Heritability estimates for DKD, CKD, and DM are presented in Table 7.
Table 4
cis-eQTLs of SNPs associated with DKD
Chr
SNP
Gene symbol
P
NES
Tissue
6
rs3128852
TRIM27
2.10×10−9
−0.19
Whole blood
HLA-A
1.60×10−7
0.23
Adipose - Subcutaneous
9
rs117744700
-
-
-
-
6
rs28366355
CYP21A1P
1.30E−11
0.30
Whole blood
1.50E−08
0.28
Artery - Tibial
HLA-DQA1
5.40E−15
−0.34
Artery - Aorta
5.00E−12
−0.22
Artery - Tibial
6.20E−11
−0.27
Adipose - Subcutaneous
6.60E−09
−0.13
Whole blood
HLA-DQA2
1.00E−97
0.90
Whole blood
6.70E−80
1.00
Adipose - Subcutaneous
2.30E−77
0.91
Artery - Tibial
8.50E−45
0.93
Heart - Left ventricle
HLA-DQB1
4.30E−13
−0.25
Whole blood
2.90E−11
−0.34
Adipose - Subcutaneous
8.30E−08
−0.34
Heart - Left ventricle
2.00E−07
−0.22
Artery - Tibial
2.00E−07
−0.28
Artery - Aorta
HLA-DQB2
2.00E−24
0.44
Whole blood
1.90E−15
0.38
Artery - Tibial
8.10E−15
0.46
Adipose - Subcutaneous
HLA-DRB1
1.70E−25
−0.18
Whole blood
1.90E−22
−0.24
Artery - Tibial
5.20E−22
−0.37
Heart - Left ventricle
1.20E−21
−0.32
Adipose - Subcutaneous
4.70E−19
−0.31
Artery - Aorta
HLA-DRB6
2.40E−95
1.00
Adipose - Subcutaneous
4.70E−75
0.7
Whole blood
1.20E−67
0.67
Artery - Tibial
3.30E−41
0.71
Artery - Aorta
3.20E−16
0.34
Whole blood
XXbac-BPG154L12.4
5.30E−10
0.34
Adipose - Subcutaneous
Chr Chromosome, NES Normalized effect size
Table 5
Differentially expressed genes of significant loci for DKD using Gene Expression Omnibus (GEO) dataset (GSE30529)
Gene symbol
log FC
AveExpr
P-value
B
TRIM27
−0.506
0.066
4.10×10−3
−0.556
HLA-A
−1.053
0.071
1.19×10−3
−0.904
CYP21A1P
0.162
0.013
0.261
−5.750
HLA-DQA1
−1.758
0.951
8.09×10−3
−2.783
HLA-DQA2
NA
NA
NA
NA
HLA-DQB1
−1.234
0.307
4.78×10−4
−0.132
HLA-DQB2
−0.450
0.170
2.21×10−2
−3.698
HLA-DRB1
NA
NA
NA
NA
HLA-DRB6
−0.725
0.018
3.53×10−5
2.347
XXbac-BPG154L12.4
NA
NA
NA
NA
logFC estimate of the log2-fold-change corresponding to the effect or contrast, AveExpr average log2-expression for the probe over all arrays and channels, B log-odds that the gene is differentially expressed, NA not available
Table 6
KEGG pathway analysis results with the five genes
KEGG ID
Pathway description
Counts in network
Strength
FDR
Matching proteins in the network
hsa04612
Antigen processing and presentation
2/63
2.09
0.011
HLA-DQA1, HLA-A
hsa04940
Type I diabetes mellitus
2/39
2.30
0.011
HLA-DQA1, HLA-A
hsa05320
Autoimmune thyroid disease
2/48
2.21
0.011
HLA-DQA1, HLA-A
hsa05330
Allograft rejection
2/34
2.36
0.011
HLA-DQA1, HLA-A
hsa05332
Graft-versus-host disease
2/36
2.34
0.011
HLA-DQA1, HLA-A
hsa05416
Viral myocarditis
2/55
2.15
0.011
HLA-DQA1, HLA-A
hsa04145
Phagosome
2/142
1.74
0.024
HLA-DQA1, HLA-A
hsa04514
Cell adhesion molecules
2/137
1.76
0.024
HLA-DQA1, HLA-A
hsa05169
Epstein-Barr virus infection
2/193
1.61
0.036
HLA-DQA1, HLA-A
hsa05166
Human T cell leukemia virus 1 infection
2/211
1.57
0.039
HLA-DQA1, HLA-A
This measure describes how large the enrichment effect is. This measure describes how large the enrichment effect is. It is the ratio between the number of proteins in the network that are annotated with a term and the number of proteins that we can expect to be annotated with this term in a random network of the same size
KEGG Kyoto Encyclopedia of Genes and Genomes, FDR false discovery rate, Strength Log10(observed/expected)
Table 7
Heritability estimates for phenotypes via GCTA
 
DM
CKD
DKD
V(G)/Vp
0.176
0.054
0.064
SE
0.012
0.011
0.011
LRT
36.867
1.488
6.437
P
6.32×10−10
0.111
5.58×10−3
GCTA Genome-wide complex trait analysis, V(G)/Vp ratio of genetic variance to phenotypic variance, SE standard error, LRT likelihood ratio test, P P-value, DM diabetes mellitus, CKD chronic kidney disease, DKD diabetic kidney disease

Transcriptome-wide association analysis (TWAS) for DKD

mRNA expression levels for 7254 protein-coding genes were imputed for TWAS. Additional file 2: Fig. S6 presents the Q-Q plot, which verified that there was no inflation in the test statistics (VIF=1.047), and volcano plots of the results of the TWAS. None of the genes showed significant differences between the patients with DKD and the control group in the DEG analysis. Summary statistics for TWAS are provided in Additional file 1: Table S7.

Discussion

In this study, we demonstrated that three novel SNPs (rs3128852, rs117744700, and rs28366355) are significantly linked to DKD. In particular, we noted that the potential causal relationship between rs1328852 and DKD was also confirmed through fine-mapping analysis. The functional analysis for rs3128852 suggests that TRIM27 and HLA-A are potential genes for determining DKD pathophysiology. This study has elucidated the pathological mechanism of DKD through genome analysis.
Earlier, GWASs for DKD or CKD were limited to the analysis of specific phenotypes, such as uric acid, eGFR, ESRD, and proteinuria [1119], and several key genes were identified—UMOD, MANBA, DAB2, and SHROOM3 [50]. We hypothesized that comparing the genomes of patients with DKD and healthy normal individuals would reveal DM-related SNPs and CKD-related SNPs; hence, we divided our investigation into four subgroups, which were specialized for DKD phenotypes. In this study, the eQTL for rs3128852 showed substantial TRIM27 and HLA-A expression, and the results of the subsequent functional genome studies supported these results.
TRIM27 encodes the tripartite motif protein family, which is involved in a variety of biological activities that may be related to autophagy and pyroptosis [51]. Recent studies demonstrated that TRIM27 was involved in the injury of glomerular endothelial cells in lupus nephritis (LN) through the FoxO1 pathway [52, 53] and in IgA nephropathy (IgAN) via T cell signaling [54]. Although the pathogenic processes of DKD and immune-related neuropathy such as LN and IgAN are different, the molecular pathways in cells may overlap, which supports our previous findings that suppression of the protein kinase B pathway could attenuate the damage by mediating the expression of TRIM27 [52, 55]. Autophagy is strictly regulated to maintain an optimal balance of cellular component synthesis, degradation, utilization, and recycling of cellular components [56]. When kidney cells are exposed to stress, dysregulated autophagy may contribute to the accumulation of cellular damage, resulting in age-related kidney disease [56]. Several experimental studies have shown that autophagy is inhibited by podocytes or proximal tubule epithelial cells [5759], which is consistent with our results. The accumulation of mitochondria plays a key role in the formation of reactive oxygen species, which activates pro-apoptotic signals and may result in hypertrophy of podocytes [60, 61], apoptosis of proximal tubular cells, and kidney fibrosis caused by the WNT-inducible signaling protein-1 [62]. Moreover, upregulation of nephrin expression in the glomeruli inhibits the expression of mammalian target rapamycin, which promotes progressive tubular damage [63]. These results support the notion that profound autophagy dysregulation is related to DKD [64].
Our study discovered that HLA-A-related genes (HLA-A, HLA-DQA1, HLA-DQB1, HLA-DQB2, and HLA-DRB6) were involved in the etiology of DKD. Although research has been conducted on the role of the immune system in CKD development, there are few studies on DKD; therefore, these results must be taken into account. According to previous studies [65, 66], renal function is associated with HLA type, such as HLA-A*01:01, HLA-A*03:01, and DQB1*02:01, which were related to CKD or ESRD. Furthermore, immune mechanisms may play a crucial role in DKD pathogenesis, especially leukocyte accumulation and associated molecular mechanisms [67]. Moreover, hyperglycemia-induced oxidative stress pathologically stimulates circulating immune cells, which enter the affected kidney and exacerbate tissue inflammation by producing pro-inflammatory cytokines and chemokines abundantly [68]. Furthermore, DNA methylation via the upregulated activity of DNA methyltransferase 1 revealed that inflamed memory immune cells aggravate DKD [54]. In addition, HLA-DPA1 may be involved in immune mechanisms underlying DKD development, since it has been identified as a significant gene for DKD [69]. Nevertheless, inflammatory response is a major factor in the progression of DKD, and the immune response exacerbates inflammation, indicating that the adaptive immune response is crucial in DKD [70, 71]. Combining the relevance of immune responses in DKD and the results of this study, immune responses and autophagy may be considered as possible pathways in the pathophysiology of DKD.
The true strength of this study includes the utilization of a relatively large elderly cohort sample that provides a better DKD phenotype. This is because diabetes is an age-related disorder, and a longer duration of diabetes is connected with an increased risk of developing DKD [24]. Furthermore, our study focused on DKD using MLR and compared it with DM without CKD and CKD without DM, which have not been evaluated earlier. However, there are a few limitations of this study. First, in this study, DKD was not diagnosed through kidney biopsy, but by clinical diagnosis. In a previous genetic study of patients with DKD selected based on a definite diagnosis via a kidney biopsy [72], it was difficult to recruit a significant number of participants due to the diversity of the etiological processes of DKD, limiting the study methodology. Moreover, diagnostic kidney biopsy is rarely performed in clinical practice [73]. However, our work has overcome this limitation by using in silico analysis and generating reproducible results, minimizing this disadvantage. Second, subjects with T1D would be at greater risk of developing DKD due to longer DM duration, and the genome-wide significant SNPs can have a vertical or horizontal pleiotropic effect on T1D and DKD. For instance, rs28366355, located near HLA, was significantly associated with DKD in our analysis; this significant result can be inferred from its association with T1D. However, for rs3128852 and rs117744700 or nearby SNPs, no significant results were reported and pleiotropic effects thereof on T1D and DKD are not expected. Furthermore, the prevalence of T1D is very rare, ranging from 0.017 to 0.021% in Koreans [74]. Thus, most individuals with DKD in our analyses may not experience T1D, and there is very low chance of a confounding effect by T1D. Further studies with individuals with DKD and T1D and T2D disease status are necessary. Third, as a study design, a mega-analysis was conducted; however, the disparity in the cohort mix is limiting. Since the CAVAS cohort was established for cardiovascular disease research, it has relatively high prevalence of hypertension and non-DM-CKD. Furthermore, the VHSMC cohort is a hospital-based cohort, whereas the KoGES consortium is based on community survey results. Hence, the severity of diabetes in these cohorts is different. By contrast, the difference in diabetes severity across these cohorts might yield more relevant results when analyzed with respect to real-world data. Fourth, bioinformatics analysis revealed that certain genetic variants and metabolic pathways were related to DKD pathogenesis, but the underlying mechanism of these factors needs further investigation. Because a simple overlap of GWAS lead variants with GTEx nominal P-value results is expected to yield several false-positive candidate causal genes [75], we conducted a QTL colocalization analysis and TWAS. However, the results were underpowered given the sample size in our study. In the PheWAS catalog (https://​phewascatalog.​org/​), for the SNP located in HLA-A (rs2860580) and its LD relationship with the top SNP (rs3128852), there is a significant association with genito-urinary phenotype (Additional file 2: Fig. S7). Further studies are required to elucidate the mechanism through which immune responses and autophagy influence DKD pathogenesis, as discovered in our study. Fifth, our study results are related to some HLA-related regions, and typically, the HLA region has a higher level of variability than the rest of the genome; thus, we need to be careful when interpreting the results. To address these concerns, we attempted to address this issue by applying an MDS plot and conducting a fine-mapping analysis. Nevertheless, the variants identified as significant genome-wide in our study were not all fine-mapped because fine-mapping analysis requires high-quality genetic data and a much larger sample size than that required for a GWAS [76].

Conclusions

This study has demonstrated that three novel SNPs (rs3128852, rs117744700, and rs28366355) are significantly associated with DKD based on the MLR GWAS mega-analysis. Moreover, the causal relationship between rs1328852 and DKD was confirmed through fine-mapping analysis. The functional analysis of the genetic variants (rs1328852) detected has revealed that TRIM27 and HLA-A, associated with immune response and autophagy, contribute to the etiology of DKD. Considering the mechanism through which immune responses and autophagy influence the pathophysiology of DKD, further research is necessary for effective prevention and treatment of DKD.

Acknowledgements

This study was conducted using the bioresources from the National Biobank of Korea, the Korea Disease Control and Prevention Agency, Republic of Korea (KBN-2021-042).
Korean Genome and Epidemiology Study (KoGES) consortium was used to obtain the following three population-based cohorts.
- Korean Association Resource from Ansan and Ansung (KARE, KoGES Ansan and Ansung study)
- Health Examinees (HEXA) cohort (KoGES Health Examinee study)
- Cardiovascular disease association (CAVAS) cohort (KoGES cardiovascular disease association study)
Veterans Health Service Medical Center cohort (VHSMC cohort)
Rex Soft provided technical support for the Korean-Chip data quality control and imputation analysis.
Genotype-Tissue Expression (GTEx) dataset: (https://​gtexportal.​org/​home/​)
Search Tool for the Retrieval of Interacting Genes/Proteins (STRING): (https://​string-db.​org/​)
Kyoto Encyclopedia of Genes and Genomes (KEGG) Database project: (https://​www.​genome.​jp/​kegg/​)

Declarations

The institutional review board of the Veterans Health Service Medical Center approved the study protocol for VHSMC cohorts after obtaining informed consent (IRB No.2018-08-032 and IRB No. 2020-02-031). The IRB approved the protocol for the KoGES consortium (IRB No. 2021-05-007) after waiving the need for informed consent since this was a retrospective study and anonymized and de-identified. Informed consent of the participants in the primary KoGES consortium had been obtained.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, et al. Diabetic kidney disease: a report from an ADA Consensus Conference. Diabetes Care. 2014;37(10):2864–83.CrossRef Tuttle KR, Bakris GL, Bilous RW, Chiang JL, de Boer IH, Goldstein-Fuchs J, et al. Diabetic kidney disease: a report from an ADA Consensus Conference. Diabetes Care. 2014;37(10):2864–83.CrossRef
2.
Zurück zum Zitat Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382(9888):260–72.CrossRef Jha V, Garcia-Garcia G, Iseki K, Li Z, Naicker S, Plattner B, et al. Chronic kidney disease: global dimension and perspectives. Lancet. 2013;382(9888):260–72.CrossRef
3.
Zurück zum Zitat Chatzikyrkou C, Menne J, Izzo J, Viberti G, Rabelink T, Ruilope LM, et al. Predictors for the development of microalbuminuria and interaction with renal function. J Hypertens. 2017;35(12):2501–9.CrossRef Chatzikyrkou C, Menne J, Izzo J, Viberti G, Rabelink T, Ruilope LM, et al. Predictors for the development of microalbuminuria and interaction with renal function. J Hypertens. 2017;35(12):2501–9.CrossRef
4.
Zurück zum Zitat Skupien J, Warram JH, Smiles A, Galecki A, Stanton RC, Krolewski AS. Improved glycemic control and risk of ESRD in patients with type 1 diabetes and proteinuria. J Am Soc Nephrol. 2014;25(12):2916–25.CrossRef Skupien J, Warram JH, Smiles A, Galecki A, Stanton RC, Krolewski AS. Improved glycemic control and risk of ESRD in patients with type 1 diabetes and proteinuria. J Am Soc Nephrol. 2014;25(12):2916–25.CrossRef
5.
Zurück zum Zitat Fioretto P, Barzon I, Mauer M. Is diabetic nephropathy reversible? Diabetes Res Clin Pract. 2014;104(3):323–8.CrossRef Fioretto P, Barzon I, Mauer M. Is diabetic nephropathy reversible? Diabetes Res Clin Pract. 2014;104(3):323–8.CrossRef
6.
Zurück zum Zitat Wiseman MJ, Saunders AJ, Keen H, Viberti G. Effect of blood glucose control on increased glomerular filtration rate and kidney size in insulin-dependent diabetes. N Engl J Med. 1985;312(10):617–21.CrossRef Wiseman MJ, Saunders AJ, Keen H, Viberti G. Effect of blood glucose control on increased glomerular filtration rate and kidney size in insulin-dependent diabetes. N Engl J Med. 1985;312(10):617–21.CrossRef
7.
Zurück zum Zitat Krolewski AS, Skupien J, Rossing P, Warram JH. Fast renal decline to end-stage renal disease: an unrecognized feature of nephropathy in diabetes. Kidney Int. 2017;91(6):1300–11.CrossRef Krolewski AS, Skupien J, Rossing P, Warram JH. Fast renal decline to end-stage renal disease: an unrecognized feature of nephropathy in diabetes. Kidney Int. 2017;91(6):1300–11.CrossRef
8.
Zurück zum Zitat Seaquist ER, Goetz FC, Rich S, Barbosa J. Familial clustering of diabetic kidney disease. Evidence for genetic susceptibility to diabetic nephropathy. N Engl J Med. 1989;320(18):1161–5.CrossRef Seaquist ER, Goetz FC, Rich S, Barbosa J. Familial clustering of diabetic kidney disease. Evidence for genetic susceptibility to diabetic nephropathy. N Engl J Med. 1989;320(18):1161–5.CrossRef
9.
Zurück zum Zitat Freedman BI, Spray BJ, Tuttle AB, Buckalew VM Jr. The familial risk of end-stage renal disease in African Americans. Am J Kidney Dis. 1993;21(4):387–93.CrossRef Freedman BI, Spray BJ, Tuttle AB, Buckalew VM Jr. The familial risk of end-stage renal disease in African Americans. Am J Kidney Dis. 1993;21(4):387–93.CrossRef
10.
Zurück zum Zitat Pettitt DJ, Saad MF, Bennett PH, Nelson RG, Knowler WC. Familial predisposition to renal disease in two generations of Pima Indians with type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia. 1990;33(7):438–43.CrossRef Pettitt DJ, Saad MF, Bennett PH, Nelson RG, Knowler WC. Familial predisposition to renal disease in two generations of Pima Indians with type 2 (non-insulin-dependent) diabetes mellitus. Diabetologia. 1990;33(7):438–43.CrossRef
11.
Zurück zum Zitat Sakaue S, Kanai M, Tanigawa Y, Karjalainen J, Kurki M, Koshiba S, et al. A cross-population atlas of genetic associations for 220 human phenotypes. Nat Genet. 2021;53(10):1415–24.CrossRef Sakaue S, Kanai M, Tanigawa Y, Karjalainen J, Kurki M, Koshiba S, et al. A cross-population atlas of genetic associations for 220 human phenotypes. Nat Genet. 2021;53(10):1415–24.CrossRef
12.
Zurück zum Zitat van Zuydam NR, Ahlqvist E, Sandholm N, Deshmukh H, Rayner NW, Abdalla M, et al. A genome-wide association study of diabetic kidney disease in subjects with type 2 diabetes. Diabetes. 2018;67(7):1414–27.CrossRef van Zuydam NR, Ahlqvist E, Sandholm N, Deshmukh H, Rayner NW, Abdalla M, et al. A genome-wide association study of diabetic kidney disease in subjects with type 2 diabetes. Diabetes. 2018;67(7):1414–27.CrossRef
13.
Zurück zum Zitat Guan M, Keaton JM, Dimitrov L, Hicks PJ, Xu J, Palmer ND, et al. Genome-wide association study identifies novel loci for type 2 diabetes-attributed end-stage kidney disease in African Americans. Hum Genomics. 2019;13(1):21.CrossRef Guan M, Keaton JM, Dimitrov L, Hicks PJ, Xu J, Palmer ND, et al. Genome-wide association study identifies novel loci for type 2 diabetes-attributed end-stage kidney disease in African Americans. Hum Genomics. 2019;13(1):21.CrossRef
14.
Zurück zum Zitat Iyengar SK, Sedor JR, Freedman BI, Kao WH, Kretzler M, Keller BJ, et al. Genome-wide association and trans-ethnic meta-analysis for advanced diabetic kidney disease: Family Investigation of Nephropathy and Diabetes (FIND). PLoS Genet. 2015;11(8):e1005352.CrossRef Iyengar SK, Sedor JR, Freedman BI, Kao WH, Kretzler M, Keller BJ, et al. Genome-wide association and trans-ethnic meta-analysis for advanced diabetic kidney disease: Family Investigation of Nephropathy and Diabetes (FIND). PLoS Genet. 2015;11(8):e1005352.CrossRef
15.
Zurück zum Zitat McDonough CW, Palmer ND, Hicks PJ, Roh BH, An SS, Cooke JN, et al. A genome-wide association study for diabetic nephropathy genes in African Americans. Kidney Int. 2011;79(5):563–72.CrossRef McDonough CW, Palmer ND, Hicks PJ, Roh BH, An SS, Cooke JN, et al. A genome-wide association study for diabetic nephropathy genes in African Americans. Kidney Int. 2011;79(5):563–72.CrossRef
16.
Zurück zum Zitat Germain M, Pezzolesi MG, Sandholm N, McKnight AJ, Susztak K, Lajer M, et al. SORBS1 gene, a new candidate for diabetic nephropathy: results from a multi-stage genome-wide association study in patients with type 1 diabetes. Diabetologia. 2015;58(3):543–8.CrossRef Germain M, Pezzolesi MG, Sandholm N, McKnight AJ, Susztak K, Lajer M, et al. SORBS1 gene, a new candidate for diabetic nephropathy: results from a multi-stage genome-wide association study in patients with type 1 diabetes. Diabetologia. 2015;58(3):543–8.CrossRef
17.
Zurück zum Zitat Taira M, Imamura M, Takahashi A, Kamatani Y, Yamauchi T, Araki SI, et al. A variant within the FTO confers susceptibility to diabetic nephropathy in Japanese patients with type 2 diabetes. PLoS One. 2018;13(12):e0208654.CrossRef Taira M, Imamura M, Takahashi A, Kamatani Y, Yamauchi T, Araki SI, et al. A variant within the FTO confers susceptibility to diabetic nephropathy in Japanese patients with type 2 diabetes. PLoS One. 2018;13(12):e0208654.CrossRef
18.
Zurück zum Zitat Liao LN, Chen CC, Wu FY, Lin CC, Hsiao JH, Chang CT, et al. Identified single-nucleotide polymorphisms and haplotypes at 16q22.1 increase diabetic nephropathy risk in Han Chinese population. BMC Genet. 2014;15:113.CrossRef Liao LN, Chen CC, Wu FY, Lin CC, Hsiao JH, Chang CT, et al. Identified single-nucleotide polymorphisms and haplotypes at 16q22.1 increase diabetic nephropathy risk in Han Chinese population. BMC Genet. 2014;15:113.CrossRef
19.
Zurück zum Zitat Salem RM, Todd JN, Sandholm N, Cole JB, Chen WM, Andrews D, et al. Genome-wide association study of diabetic kidney disease highlights biology involved in glomerular basement membrane collagen. J Am Soc Nephrol. 2019;30(10):2000–16.CrossRef Salem RM, Todd JN, Sandholm N, Cole JB, Chen WM, Andrews D, et al. Genome-wide association study of diabetic kidney disease highlights biology involved in glomerular basement membrane collagen. J Am Soc Nephrol. 2019;30(10):2000–16.CrossRef
20.
Zurück zum Zitat Gu HF. Genetic and epigenetic studies in diabetic kidney disease. Front Genet. 2019;10:507.CrossRef Gu HF. Genetic and epigenetic studies in diabetic kidney disease. Front Genet. 2019;10:507.CrossRef
21.
Zurück zum Zitat Mohamed SA, Fernadez-Tajes J, Franks PW, Bennet L. GWAS in people of Middle Eastern descent reveals a locus protective of kidney function-a cross-sectional study. BMC Med. 2022;20(1):76.CrossRef Mohamed SA, Fernadez-Tajes J, Franks PW, Bennet L. GWAS in people of Middle Eastern descent reveals a locus protective of kidney function-a cross-sectional study. BMC Med. 2022;20(1):76.CrossRef
22.
Zurück zum Zitat Ku E, Lee BJ, Wei J, Weir MR. Hypertension in CKD: Core Curriculum 2019. Am J Kidney Dis. 2019;74(1):120–31.CrossRef Ku E, Lee BJ, Wei J, Weir MR. Hypertension in CKD: Core Curriculum 2019. Am J Kidney Dis. 2019;74(1):120–31.CrossRef
23.
Zurück zum Zitat Control CfD, Prevention. National diabetes fact sheet: general information and national estimates on diabetes in the United State. Atlanta: US Department of Health and Human Services, Centers for Disease Control and Prevention; 2007. Control CfD, Prevention. National diabetes fact sheet: general information and national estimates on diabetes in the United State. Atlanta: US Department of Health and Human Services, Centers for Disease Control and Prevention; 2007.
24.
Zurück zum Zitat Anders HJ, Huber TB, Isermann B, Schiffer M. CKD in diabetes: diabetic kidney disease versus nondiabetic kidney disease. Nat Rev Nephrol. 2018;14(6):361–77.CrossRef Anders HJ, Huber TB, Isermann B, Schiffer M. CKD in diabetes: diabetic kidney disease versus nondiabetic kidney disease. Nat Rev Nephrol. 2018;14(6):361–77.CrossRef
25.
Zurück zum Zitat Ahluwalia TS, Lindholm E, Groop L, Melander O. Uromodulin gene variant is associated with type 2 diabetic nephropathy. J Hypertens. 2011;29(9):1731–4.CrossRef Ahluwalia TS, Lindholm E, Groop L, Melander O. Uromodulin gene variant is associated with type 2 diabetic nephropathy. J Hypertens. 2011;29(9):1731–4.CrossRef
26.
Zurück zum Zitat Prudente S, Di Paola R, Copetti M, Lucchesi D, Lamacchia O, Pezzilli S, et al. The rs12917707 polymorphism at the UMOD locus and glomerular filtration rate in individuals with type 2 diabetes: evidence of heterogeneity across two different European populations. Nephrol Dial Transplant. 2017;32(10):1718–22. Prudente S, Di Paola R, Copetti M, Lucchesi D, Lamacchia O, Pezzilli S, et al. The rs12917707 polymorphism at the UMOD locus and glomerular filtration rate in individuals with type 2 diabetes: evidence of heterogeneity across two different European populations. Nephrol Dial Transplant. 2017;32(10):1718–22.
27.
Zurück zum Zitat Maeda S, Kobayashi MA, Araki S, Babazono T, Freedman BI, Bostrom MA, et al. A single nucleotide polymorphism within the acetyl-coenzyme A carboxylase beta gene is associated with proteinuria in patients with type 2 diabetes. PLoS Genet. 2010;6(2):e1000842.CrossRef Maeda S, Kobayashi MA, Araki S, Babazono T, Freedman BI, Bostrom MA, et al. A single nucleotide polymorphism within the acetyl-coenzyme A carboxylase beta gene is associated with proteinuria in patients with type 2 diabetes. PLoS Genet. 2010;6(2):e1000842.CrossRef
28.
Zurück zum Zitat Kim YA, Lee Y, Seo JH. Renal complication and glycemic control in Korean veterans with type 2 diabetes: a 10-year retrospective cohort study. J Diabetes Res. 2020;2020:9806790.CrossRef Kim YA, Lee Y, Seo JH. Renal complication and glycemic control in Korean veterans with type 2 diabetes: a 10-year retrospective cohort study. J Diabetes Res. 2020;2020:9806790.CrossRef
29.
Zurück zum Zitat Lee JH, Kim YA, Lee Y, Bang WD, Seo JH. Association between interarm blood pressure differences and diabetic retinopathy in patients with type 2 diabetes. Diab Vasc Dis Res. 2020;17(7):1479164120945910. Lee JH, Kim YA, Lee Y, Bang WD, Seo JH. Association between interarm blood pressure differences and diabetic retinopathy in patients with type 2 diabetes. Diab Vasc Dis Res. 2020;17(7):1479164120945910.
30.
Zurück zum Zitat Kim Y, Han BG, Ko GESg. Cohort profile: the Korean genome and epidemiology study (KoGES) consortium. Int J Epidemiol. 2017;46(2):e20.CrossRef Kim Y, Han BG, Ko GESg. Cohort profile: the Korean genome and epidemiology study (KoGES) consortium. Int J Epidemiol. 2017;46(2):e20.CrossRef
31.
Zurück zum Zitat Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247–54.CrossRef Levey AS, Coresh J, Greene T, Stevens LA, Zhang YL, Hendriksen S, et al. Using standardized serum creatinine values in the modification of diet in renal disease study equation for estimating glomerular filtration rate. Ann Intern Med. 2006;145(4):247–54.CrossRef
32.
Zurück zum Zitat Inker LA, Astor BC, Fox CH, Isakova T, Lash JP, Peralta CA, et al. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for the evaluation and management of CKD. Am J Kidney Dis. 2014;63(5):713–35.CrossRef Inker LA, Astor BC, Fox CH, Isakova T, Lash JP, Peralta CA, et al. KDOQI US commentary on the 2012 KDIGO clinical practice guideline for the evaluation and management of CKD. Am J Kidney Dis. 2014;63(5):713–35.CrossRef
33.
Zurück zum Zitat Moon S, Kim YJ, Han S, Hwang MY, Shin DM, Park MY, et al. The Korea Biobank Array: design and identification of coding variants associated with blood biochemical traits. Sci Rep. 2019;9(1):1382.CrossRef Moon S, Kim YJ, Han S, Hwang MY, Shin DM, Park MY, et al. The Korea Biobank Array: design and identification of coding variants associated with blood biochemical traits. Sci Rep. 2019;9(1):1382.CrossRef
34.
Zurück zum Zitat Seo S, Park K, Lee JJ, Choi KY, Lee KH, Won S. SNP genotype calling and quality control for multi-batch-based studies. Genes Genomics. 2019;41(8):927–39.CrossRef Seo S, Park K, Lee JJ, Choi KY, Lee KH, Won S. SNP genotype calling and quality control for multi-batch-based studies. Genes Genomics. 2019;41(8):927–39.CrossRef
35.
Zurück zum Zitat Song YE, Lee S, Park K, Elston RC, Yang H-J, Won S. ONETOOL for the analysis of family-based big data. Bioinformatics. 2018;34(16):2851–3.CrossRef Song YE, Lee S, Park K, Elston RC, Yang H-J, Won S. ONETOOL for the analysis of family-based big data. Bioinformatics. 2018;34(16):2851–3.CrossRef
36.
Zurück zum Zitat McCarthy S, Das S, Kretzschmar W, Delaneau O, Wood AR, Teumer A, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet. 2016;48(10):1279–83.CrossRef McCarthy S, Das S, Kretzschmar W, Delaneau O, Wood AR, Teumer A, et al. A reference panel of 64,976 haplotypes for genotype imputation. Nat Genet. 2016;48(10):1279–83.CrossRef
37.
Zurück zum Zitat Loh P-R, Danecek P, Palamara PF, Fuchsberger C, Reshef YA, Finucane HK, et al. Reference-based phasing using the haplotype reference consortium panel. Nat Genet. 2016;48(11):1443–8.CrossRef Loh P-R, Danecek P, Palamara PF, Fuchsberger C, Reshef YA, Finucane HK, et al. Reference-based phasing using the haplotype reference consortium panel. Nat Genet. 2016;48(11):1443–8.CrossRef
38.
Zurück zum Zitat Das S, Forer L, Schönherr S, Sidore C, Locke AE, Kwong A, et al. Next-generation genotype imputation service and methods. Nat Genet. 2016;48(10):1284–7.CrossRef Das S, Forer L, Schönherr S, Sidore C, Locke AE, Kwong A, et al. Next-generation genotype imputation service and methods. Nat Genet. 2016;48(10):1284–7.CrossRef
39.
Zurück zum Zitat Marchini J, Howie B, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat Genet. 2007;39(7):906–13.CrossRef Marchini J, Howie B, Myers S, McVean G, Donnelly P. A new multipoint method for genome-wide association studies by imputation of genotypes. Nat Genet. 2007;39(7):906–13.CrossRef
40.
Zurück zum Zitat Pruim RJ, Welch RP, Sanna S, Teslovich TM, Chines PS, Gliedt TP, et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010;26(18):2336–7.CrossRef Pruim RJ, Welch RP, Sanna S, Teslovich TM, Chines PS, Gliedt TP, et al. LocusZoom: regional visualization of genome-wide association scan results. Bioinformatics. 2010;26(18):2336–7.CrossRef
41.
Zurück zum Zitat Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet. 2011;88(1):76–82.CrossRef Yang J, Lee SH, Goddard ME, Visscher PM. GCTA: a tool for genome-wide complex trait analysis. Am J Hum Genet. 2011;88(1):76–82.CrossRef
42.
Zurück zum Zitat Sharp SA, Rich SS, Wood AR, Jones SE, Beaumont RN, Harrison JW, et al. Development and standardization of an improved type 1 diabetes genetic risk score for use in newborn screening and incident diagnosis. Diabetes Care. 2019;42(2):200–7.CrossRef Sharp SA, Rich SS, Wood AR, Jones SE, Beaumont RN, Harrison JW, et al. Development and standardization of an improved type 1 diabetes genetic risk score for use in newborn screening and incident diagnosis. Diabetes Care. 2019;42(2):200–7.CrossRef
43.
Zurück zum Zitat Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81(3):559–75.CrossRef Purcell S, Neale B, Todd-Brown K, Thomas L, Ferreira MA, Bender D, et al. PLINK: a tool set for whole-genome association and population-based linkage analyses. Am J Hum Genet. 2007;81(3):559–75.CrossRef
44.
Zurück zum Zitat Kichaev G, Yang W-Y, Lindstrom S, Hormozdiari F, Eskin E, Price AL, et al. Integrating functional data to prioritize causal variants in statistical fine-mapping studies. PLoS Genet. 2014;10(10):e1004722.CrossRef Kichaev G, Yang W-Y, Lindstrom S, Hormozdiari F, Eskin E, Price AL, et al. Integrating functional data to prioritize causal variants in statistical fine-mapping studies. PLoS Genet. 2014;10(10):e1004722.CrossRef
45.
Zurück zum Zitat Lonsdale J, Thomas J, Salvatore M, Phillips R, Lo E, Shad S, et al. The genotype-tissue expression (GTEx) project. Nat Genet. 2013;45(6):580–5.CrossRef Lonsdale J, Thomas J, Salvatore M, Phillips R, Lo E, Shad S, et al. The genotype-tissue expression (GTEx) project. Nat Genet. 2013;45(6):580–5.CrossRef
46.
Zurück zum Zitat Kelly-Smith M, Strain GM. STRING data mining of GWAS data in canine hereditary pigment-associated deafness. Vet Anim Sci. 2020;9:100118.CrossRef Kelly-Smith M, Strain GM. STRING data mining of GWAS data in canine hereditary pigment-associated deafness. Vet Anim Sci. 2020;9:100118.CrossRef
47.
Zurück zum Zitat Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–51.CrossRef Kanehisa M, Furumichi M, Sato Y, Ishiguro-Watanabe M, Tanabe M. KEGG: integrating viruses and cellular organisms. Nucleic Acids Res. 2021;49(D1):D545–51.CrossRef
48.
Zurück zum Zitat Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K, Carroll RJ, et al. A gene-based association method for mapping traits using reference transcriptome data. Nat Genet. 2015;47(9):1091–8.CrossRef Gamazon ER, Wheeler HE, Shah KP, Mozaffari SV, Aquino-Michaels K, Carroll RJ, et al. A gene-based association method for mapping traits using reference transcriptome data. Nat Genet. 2015;47(9):1091–8.CrossRef
49.
Zurück zum Zitat Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.CrossRef Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47.CrossRef
50.
Zurück zum Zitat Sullivan KM, Susztak K. Unravelling the complex genetics of common kidney diseases: from variants to mechanisms. Nat Rev Nephrol. 2020;16(11):628–40.CrossRef Sullivan KM, Susztak K. Unravelling the complex genetics of common kidney diseases: from variants to mechanisms. Nat Rev Nephrol. 2020;16(11):628–40.CrossRef
51.
Zurück zum Zitat Wan T, Li X, Li Y. The role of TRIM family proteins in autophagy, pyroptosis, and diabetes mellitus. Cell Biol Int. 2021;45(5):913–26.CrossRef Wan T, Li X, Li Y. The role of TRIM family proteins in autophagy, pyroptosis, and diabetes mellitus. Cell Biol Int. 2021;45(5):913–26.CrossRef
52.
Zurück zum Zitat Liu J, Xu J, Huang J, Gu C, Liu Q, Zhang W, et al. TRIM27 contributes to glomerular endothelial cell injury in lupus nephritis by mediating the FoxO1 signaling pathway. Lab Invest. 2021;101(8):983–97.CrossRef Liu J, Xu J, Huang J, Gu C, Liu Q, Zhang W, et al. TRIM27 contributes to glomerular endothelial cell injury in lupus nephritis by mediating the FoxO1 signaling pathway. Lab Invest. 2021;101(8):983–97.CrossRef
53.
Zurück zum Zitat Liu J, Feng X, Tian Y, Wang K, Gao F, Yang L, et al. Knockdown of TRIM27 expression suppresses the dysfunction of mesangial cells in lupus nephritis by FoxO1 pathway. J Cell Physiol. 2019;234(7):11555–66.CrossRef Liu J, Feng X, Tian Y, Wang K, Gao F, Yang L, et al. Knockdown of TRIM27 expression suppresses the dysfunction of mesangial cells in lupus nephritis by FoxO1 pathway. J Cell Physiol. 2019;234(7):11555–66.CrossRef
54.
Zurück zum Zitat Chen XJ, Zhang H, Yang F, Liu Y, Chen G. DNA methylation sustains “inflamed” memory of peripheral immune cells aggravating kidney inflammatory response in chronic kidney disease. Front Physiol. 2021;12:637480.CrossRef Chen XJ, Zhang H, Yang F, Liu Y, Chen G. DNA methylation sustains “inflamed” memory of peripheral immune cells aggravating kidney inflammatory response in chronic kidney disease. Front Physiol. 2021;12:637480.CrossRef
55.
Zurück zum Zitat Ren H, Shao Y, Wu C, Ma X, Lv C, Wang Q. Metformin alleviates oxidative stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1 pathway. Mol Cell Endocrinol. 2020;500:110628.CrossRef Ren H, Shao Y, Wu C, Ma X, Lv C, Wang Q. Metformin alleviates oxidative stress and enhances autophagy in diabetic kidney disease via AMPK/SIRT1-FoxO1 pathway. Mol Cell Endocrinol. 2020;500:110628.CrossRef
56.
Zurück zum Zitat Yang D, Livingston MJ, Liu Z, Dong G, Zhang M, Chen JK, et al. Autophagy in diabetic kidney disease: regulation, pathological role and therapeutic potential. Cell Mol Life Sci. 2018;75(4):669–88.CrossRef Yang D, Livingston MJ, Liu Z, Dong G, Zhang M, Chen JK, et al. Autophagy in diabetic kidney disease: regulation, pathological role and therapeutic potential. Cell Mol Life Sci. 2018;75(4):669–88.CrossRef
57.
Zurück zum Zitat Vallon V, Rose M, Gerasimova M, Satriano J, Platt KA, Koepsell H, et al. Knockout of Na-glucose transporter SGLT2 attenuates hyperglycemia and glomerular hyperfiltration but not kidney growth or injury in diabetes mellitus. Am J Physiol Renal Physiol. 2013;304(2):F156–67.CrossRef Vallon V, Rose M, Gerasimova M, Satriano J, Platt KA, Koepsell H, et al. Knockout of Na-glucose transporter SGLT2 attenuates hyperglycemia and glomerular hyperfiltration but not kidney growth or injury in diabetes mellitus. Am J Physiol Renal Physiol. 2013;304(2):F156–67.CrossRef
58.
Zurück zum Zitat Kitada M, Takeda A, Nagai T, Ito H, Kanasaki K, Koya D. Dietary restriction ameliorates diabetic nephropathy through anti-inflammatory effects and regulation of the autophagy via restoration of Sirt1 in diabetic Wistar fatty (fa/fa) rats: a model of type 2 diabetes. Exp Diabetes Res. 2011;2011:908185.CrossRef Kitada M, Takeda A, Nagai T, Ito H, Kanasaki K, Koya D. Dietary restriction ameliorates diabetic nephropathy through anti-inflammatory effects and regulation of the autophagy via restoration of Sirt1 in diabetic Wistar fatty (fa/fa) rats: a model of type 2 diabetes. Exp Diabetes Res. 2011;2011:908185.CrossRef
59.
Zurück zum Zitat Yamahara K, Kume S, Koya D, Tanaka Y, Morita Y, Chin-Kanasaki M, et al. Obesity-mediated autophagy insufficiency exacerbates proteinuria-induced tubulointerstitial lesions. J Am Soc Nephrol. 2013;24(11):1769–81.CrossRef Yamahara K, Kume S, Koya D, Tanaka Y, Morita Y, Chin-Kanasaki M, et al. Obesity-mediated autophagy insufficiency exacerbates proteinuria-induced tubulointerstitial lesions. J Am Soc Nephrol. 2013;24(11):1769–81.CrossRef
60.
Zurück zum Zitat Hartleben B, Godel M, Meyer-Schwesinger C, Liu S, Ulrich T, Kobler S, et al. Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. J Clin Invest. 2010;120(4):1084–96.CrossRef Hartleben B, Godel M, Meyer-Schwesinger C, Liu S, Ulrich T, Kobler S, et al. Autophagy influences glomerular disease susceptibility and maintains podocyte homeostasis in aging mice. J Clin Invest. 2010;120(4):1084–96.CrossRef
61.
Zurück zum Zitat Koch EAT, Nakhoul R, Nakhoul F, Nakhoul N. Autophagy in diabetic nephropathy: a review. Int Urol Nephrol. 2020;52(9):1705–12.CrossRef Koch EAT, Nakhoul R, Nakhoul F, Nakhoul N. Autophagy in diabetic nephropathy: a review. Int Urol Nephrol. 2020;52(9):1705–12.CrossRef
62.
Zurück zum Zitat Yang X, Wang H, Tu Y, Li Y, Zou Y, Li G, et al. WNT1-inducible signaling protein-1 mediates TGF-beta1-induced renal fibrosis in tubular epithelial cells and unilateral ureteral obstruction mouse models via autophagy. J Cell Physiol. 2020;235(3):2009–22.CrossRef Yang X, Wang H, Tu Y, Li Y, Zou Y, Li G, et al. WNT1-inducible signaling protein-1 mediates TGF-beta1-induced renal fibrosis in tubular epithelial cells and unilateral ureteral obstruction mouse models via autophagy. J Cell Physiol. 2020;235(3):2009–22.CrossRef
63.
Zurück zum Zitat Nolin AC, Mulhern RM, Panchenko MV, Pisarek-Horowitz A, Wang Z, Shirihai O, et al. Proteinuria causes dysfunctional autophagy in the proximal tubule. Am J Physiol Renal Physiol. 2016;311(6):F1271–9.CrossRef Nolin AC, Mulhern RM, Panchenko MV, Pisarek-Horowitz A, Wang Z, Shirihai O, et al. Proteinuria causes dysfunctional autophagy in the proximal tubule. Am J Physiol Renal Physiol. 2016;311(6):F1271–9.CrossRef
64.
Zurück zum Zitat Gonzalez CD, Carro Negueruela MP, Nicora Santamarina C, Resnik R, Vaccaro MI. Autophagy dysregulation in diabetic kidney disease: from pathophysiology to pharmacological interventions. Cells. 2021;10(9):2497.CrossRef Gonzalez CD, Carro Negueruela MP, Nicora Santamarina C, Resnik R, Vaccaro MI. Autophagy dysregulation in diabetic kidney disease: from pathophysiology to pharmacological interventions. Cells. 2021;10(9):2497.CrossRef
65.
Zurück zum Zitat Lowe M, Payton A, Verma A, Worthington J, Gemmell I, Hamilton P, et al. Associations between human leukocyte antigens and renal function. Sci Rep. 2021;11(1):3158.CrossRef Lowe M, Payton A, Verma A, Worthington J, Gemmell I, Hamilton P, et al. Associations between human leukocyte antigens and renal function. Sci Rep. 2021;11(1):3158.CrossRef
66.
Zurück zum Zitat Robson KJ, Ooi JD, Holdsworth SR, Rossjohn J, Kitching AR. HLA and kidney disease: from associations to mechanisms. Nat Rev Nephrol. 2018;14(10):636–55.CrossRef Robson KJ, Ooi JD, Holdsworth SR, Rossjohn J, Kitching AR. HLA and kidney disease: from associations to mechanisms. Nat Rev Nephrol. 2018;14(10):636–55.CrossRef
67.
Zurück zum Zitat Yang X, Mou S. Role of immune cells in diabetic kidney disease. Curr Gene Ther. 2017;17(6):424–33.CrossRef Yang X, Mou S. Role of immune cells in diabetic kidney disease. Curr Gene Ther. 2017;17(6):424–33.CrossRef
69.
Zurück zum Zitat Ma F, Sun T, Wu M, Wang W, Xu Z. Identification of key genes for diabetic kidney disease using biological informatics methods. Mol Med Rep. 2017;16(6):7931–8.CrossRef Ma F, Sun T, Wu M, Wang W, Xu Z. Identification of key genes for diabetic kidney disease using biological informatics methods. Mol Med Rep. 2017;16(6):7931–8.CrossRef
70.
Zurück zum Zitat Kong L, Andrikopoulos S, MacIsaac RJ, Mackay LK, Nikolic-Paterson DJ, Torkamani N, et al. Role of the adaptive immune system in diabetic kidney disease. J Diabetes Investig. 2022;13(2):213–26.CrossRef Kong L, Andrikopoulos S, MacIsaac RJ, Mackay LK, Nikolic-Paterson DJ, Torkamani N, et al. Role of the adaptive immune system in diabetic kidney disease. J Diabetes Investig. 2022;13(2):213–26.CrossRef
71.
Zurück zum Zitat Tang SCW, Yiu WH. Innate immunity in diabetic kidney disease. Nat Rev Nephrol. 2020;16(4):206–22.CrossRef Tang SCW, Yiu WH. Innate immunity in diabetic kidney disease. Nat Rev Nephrol. 2020;16(4):206–22.CrossRef
72.
Zurück zum Zitat Jeong KH, Kim JS, Woo JT, Rhee SY, Lee YH, Kim YG, et al. Genome-wide association study identifies new susceptibility loci for diabetic nephropathy in Korean patients with type 2 diabetes mellitus. Clin Genet. 2019;96(1):35–42.CrossRef Jeong KH, Kim JS, Woo JT, Rhee SY, Lee YH, Kim YG, et al. Genome-wide association study identifies new susceptibility loci for diabetic nephropathy in Korean patients with type 2 diabetes mellitus. Clin Genet. 2019;96(1):35–42.CrossRef
73.
Zurück zum Zitat Fiorentino M, Bolignano D, Tesar V, Pisano A, Biesen WV, Tripepi G, et al. Renal biopsy in patients with diabetes: a pooled meta-analysis of 48 studies. Nephrol Dial Transplant. 2017;32(1):97–110. Fiorentino M, Bolignano D, Tesar V, Pisano A, Biesen WV, Tripepi G, et al. Renal biopsy in patients with diabetes: a pooled meta-analysis of 48 studies. Nephrol Dial Transplant. 2017;32(1):97–110.
74.
Zurück zum Zitat Song SO, Song YD, Nam JY, Park KH, Yoon J-H, Son K-M, et al. Epidemiology of type 1 diabetes mellitus in Korea through an investigation of the national registration project of type 1 diabetes for the reimbursement of glucometer strips with additional analyses using claims data. Diabetes Metab J. 2016;40(1):35–45.CrossRef Song SO, Song YD, Nam JY, Park KH, Yoon J-H, Son K-M, et al. Epidemiology of type 1 diabetes mellitus in Korea through an investigation of the national registration project of type 1 diabetes for the reimbursement of glucometer strips with additional analyses using claims data. Diabetes Metab J. 2016;40(1):35–45.CrossRef
75.
Zurück zum Zitat Liu B, Gloudemans MJ, Rao AS, Ingelsson E, Montgomery SB. Abundant associations with gene expression complicate GWAS follow-up. Nat Genet. 2019;51(5):768–9.CrossRef Liu B, Gloudemans MJ, Rao AS, Ingelsson E, Montgomery SB. Abundant associations with gene expression complicate GWAS follow-up. Nat Genet. 2019;51(5):768–9.CrossRef
76.
Zurück zum Zitat Wang QS, Huang H. Methods for statistical fine-mapping and their applications to auto-immune diseases. Semin Immunopathol. 2022;44(1):101–13.CrossRef Wang QS, Huang H. Methods for statistical fine-mapping and their applications to auto-immune diseases. Semin Immunopathol. 2022;44(1):101–13.CrossRef
Metadaten
Titel
Identification of genetic variants associated with diabetic kidney disease in multiple Korean cohorts via a genome-wide association study mega-analysis
verfasst von
Heejin Jin
Ye An Kim
Young Lee
Seung-hyun Kwon
Ah Ra Do
Sujin Seo
Sungho Won
Je Hyun Seo
Publikationsdatum
01.12.2023
Verlag
BioMed Central
Erschienen in
BMC Medicine / Ausgabe 1/2023
Elektronische ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-022-02723-4

Weitere Artikel der Ausgabe 1/2023

BMC Medicine 1/2023 Zur Ausgabe

Leitlinien kompakt für die Allgemeinmedizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Facharzt-Training Allgemeinmedizin

Die ideale Vorbereitung zur anstehenden Prüfung mit den ersten 49 von 100 klinischen Fallbeispielen verschiedener Themenfelder

Mehr erfahren

Bei Herzinsuffizienz muss „Eisenmangel“ neu definiert werden

16.05.2024 Herzinsuffizienz Nachrichten

Bei chronischer Herzinsuffizienz macht es einem internationalen Expertenteam zufolge wenig Sinn, die Diagnose „Eisenmangel“ am Serumferritin festzumachen. Das Team schlägt vor, sich lieber an die Transferrinsättigung zu halten.

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Betalaktam-Allergie: praxisnahes Vorgehen beim Delabeling

16.05.2024 Pädiatrische Allergologie Nachrichten

Die große Mehrheit der vermeintlichen Penicillinallergien sind keine. Da das „Etikett“ Betalaktam-Allergie oft schon in der Kindheit erworben wird, kann ein frühzeitiges Delabeling lebenslange Vorteile bringen. Ein Team von Pädiaterinnen und Pädiatern aus Kanada stellt vor, wie sie dabei vorgehen.

Diabetestechnologie für alle?

15.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Eine verbesserte Stoffwechseleinstellung und höhere Lebensqualität – Diabetestechnologien sollen den Alltag der Patienten erleichtern. Dass CGM, AID & Co. bei Typ-1-Diabetes helfen, ist belegt. Bei Typ-2 gestaltet sich die Sache komplizierter.

Update Allgemeinmedizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.