Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Nat Commun ; 15(1): 586, 2024 Jan 18.
Article En | MEDLINE | ID: mdl-38233393

X-chromosomal genetic variants are understudied but can yield valuable insights into sexually dimorphic human traits and diseases. We performed a sex-stratified cross-ancestry X-chromosome-wide association meta-analysis of seven kidney-related traits (n = 908,697), identifying 23 loci genome-wide significantly associated with two of the traits: 7 for uric acid and 16 for estimated glomerular filtration rate (eGFR), including four novel eGFR loci containing the functionally plausible prioritized genes ACSL4, CLDN2, TSPAN6 and the female-specific DRP2. Further, we identified five novel sex-interactions, comprising male-specific effects at FAM9B and AR/EDA2R, and three sex-differential findings with larger genetic effect sizes in males at DCAF12L1 and MST4 and larger effect sizes in females at HPRT1. All prioritized genes in loci showing significant sex-interactions were located next to androgen response elements (ARE). Five ARE genes showed sex-differential expressions. This study contributes new insights into sex-dimorphisms of kidney traits along with new prioritized gene targets for further molecular research.


Androgens , Genome-Wide Association Study , Humans , Male , Female , Androgens/genetics , Kidney , Chromosomes, Human, X/genetics , Response Elements , Polymorphism, Single Nucleotide , Genetic Predisposition to Disease , Tetraspanins/genetics
2.
Commun Biol ; 5(1): 580, 2022 06 13.
Article En | MEDLINE | ID: mdl-35697829

Reduced glomerular filtration rate (GFR) can progress to kidney failure. Risk factors include genetics and diabetes mellitus (DM), but little is known about their interaction. We conducted genome-wide association meta-analyses for estimated GFR based on serum creatinine (eGFR), separately for individuals with or without DM (nDM = 178,691, nnoDM = 1,296,113). Our genome-wide searches identified (i) seven eGFR loci with significant DM/noDM-difference, (ii) four additional novel loci with suggestive difference and (iii) 28 further novel loci (including CUBN) by allowing for potential difference. GWAS on eGFR among DM individuals identified 2 known and 27 potentially responsible loci for diabetic kidney disease. Gene prioritization highlighted 18 genes that may inform reno-protective drug development. We highlight the existence of DM-only and noDM-only effects, which can inform about the target group, if respective genes are advanced as drug targets. Largely shared effects suggest that most drug interventions to alter eGFR should be effective in DM and noDM.


Diabetes Mellitus , Diabetic Nephropathies , Creatinine , Diabetic Nephropathies/genetics , Genome-Wide Association Study , Glomerular Filtration Rate/genetics , Humans , Kidney
3.
Nat Commun ; 12(1): 7173, 2021 12 09.
Article En | MEDLINE | ID: mdl-34887389

Elevated serum urate levels, a complex trait and major risk factor for incident gout, are correlated with cardiometabolic traits via incompletely understood mechanisms. DNA methylation in whole blood captures genetic and environmental influences and is assessed in transethnic meta-analysis of epigenome-wide association studies (EWAS) of serum urate (discovery, n = 12,474, replication, n = 5522). The 100 replicated, epigenome-wide significant (p < 1.1E-7) CpGs explain 11.6% of the serum urate variance. At SLC2A9, the serum urate locus with the largest effect in genome-wide association studies (GWAS), five CpGs are associated with SLC2A9 gene expression. Four CpGs at SLC2A9 have significant causal effects on serum urate levels and/or gout, and two of these partly mediate the effects of urate-associated GWAS variants. In other genes, including SLC7A11 and PHGDH, 17 urate-associated CpGs are associated with conditions defining metabolic syndrome, suggesting that these CpGs may represent a blood DNA methylation signature of cardiometabolic risk factors. This study demonstrates that EWAS can provide new insights into GWAS loci and the correlation of serum urate with other complex traits.


Epigenome , Glucose Transport Proteins, Facilitative/genetics , Gout/genetics , Uric Acid/blood , Amino Acid Transport System y+/genetics , Cohort Studies , CpG Islands , DNA Methylation , Female , Genetic Predisposition to Disease , Genome-Wide Association Study , Glucose Transport Proteins, Facilitative/metabolism , Gout/blood , Humans , Male
4.
Nat Commun ; 12(1): 7174, 2021 12 09.
Article En | MEDLINE | ID: mdl-34887417

Chronic kidney disease is a major public health burden. Elevated urinary albumin-to-creatinine ratio is a measure of kidney damage, and used to diagnose and stage chronic kidney disease. To extend the knowledge on regulatory mechanisms related to kidney function and disease, we conducted a blood-based epigenome-wide association study for estimated glomerular filtration rate (n = 33,605) and urinary albumin-to-creatinine ratio (n = 15,068) and detected 69 and seven CpG sites where DNA methylation was associated with the respective trait. The majority of these findings showed directionally consistent associations with the respective clinical outcomes chronic kidney disease and moderately increased albuminuria. Associations of DNA methylation with kidney function, such as CpGs at JAZF1, PELI1 and CHD2 were validated in kidney tissue. Methylation at PHRF1, LDB2, CSRNP1 and IRF5 indicated causal effects on kidney function. Enrichment analyses revealed pathways related to hemostasis and blood cell migration for estimated glomerular filtration rate, and immune cell activation and response for urinary albumin-to-creatinineratio-associated CpGs.


DNA Methylation , Renal Insufficiency, Chronic/genetics , Adult , Aged , CpG Islands , Female , Glomerular Filtration Rate , Humans , Interferon Regulatory Factors/genetics , Interferon Regulatory Factors/metabolism , Kidney/metabolism , Kidney/physiopathology , Kidney Function Tests , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Renal Insufficiency, Chronic/metabolism , Renal Insufficiency, Chronic/physiopathology , Transcription Factors/genetics , Transcription Factors/metabolism
5.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Article En | MEDLINE | ID: mdl-34548398

Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining-based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type-specific manner. These findings have implications for potential therapeutic strategies.


Bone and Bones/abnormalities , Ciliopathies/pathology , Craniosynostoses/pathology , Cytoskeletal Proteins/metabolism , Ectodermal Dysplasia/pathology , Embryo, Nonmammalian/pathology , Musculoskeletal Abnormalities/pathology , Polycystic Kidney Diseases/pathology , Tubulin/chemistry , Animals , Bone and Bones/metabolism , Bone and Bones/pathology , Ciliopathies/genetics , Ciliopathies/metabolism , Craniosynostoses/genetics , Craniosynostoses/metabolism , Cytoskeletal Proteins/genetics , Disease Models, Animal , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/metabolism , Embryo, Nonmammalian/metabolism , Musculoskeletal Abnormalities/genetics , Musculoskeletal Abnormalities/metabolism , Phenotype , Polycystic Kidney Diseases/genetics , Polycystic Kidney Diseases/metabolism , Tubulin/metabolism , Xenopus laevis
6.
Kidney Int ; 99(4): 926-939, 2021 04.
Article En | MEDLINE | ID: mdl-33137338

Rapid decline of glomerular filtration rate estimated from creatinine (eGFRcrea) is associated with severe clinical endpoints. In contrast to cross-sectionally assessed eGFRcrea, the genetic basis for rapid eGFRcrea decline is largely unknown. To help define this, we meta-analyzed 42 genome-wide association studies from the Chronic Kidney Diseases Genetics Consortium and United Kingdom Biobank to identify genetic loci for rapid eGFRcrea decline. Two definitions of eGFRcrea decline were used: 3 mL/min/1.73m2/year or more ("Rapid3"; encompassing 34,874 cases, 107,090 controls) and eGFRcrea decline 25% or more and eGFRcrea under 60 mL/min/1.73m2 at follow-up among those with eGFRcrea 60 mL/min/1.73m2 or more at baseline ("CKDi25"; encompassing 19,901 cases, 175,244 controls). Seven independent variants were identified across six loci for Rapid3 and/or CKDi25: consisting of five variants at four loci with genome-wide significance (near UMOD-PDILT (2), PRKAG2, WDR72, OR2S2) and two variants among 265 known eGFRcrea variants (near GATM, LARP4B). All these loci were novel for Rapid3 and/or CKDi25 and our bioinformatic follow-up prioritized variants and genes underneath these loci. The OR2S2 locus is novel for any eGFRcrea trait including interesting candidates. For the five genome-wide significant lead variants, we found supporting effects for annual change in blood urea nitrogen or cystatin-based eGFR, but not for GATM or LARP4B. Individuals at high compared to those at low genetic risk (8-14 vs. 0-5 adverse alleles) had a 1.20-fold increased risk of acute kidney injury (95% confidence interval 1.08-1.33). Thus, our identified loci for rapid kidney function decline may help prioritize therapeutic targets and identify mechanisms and individuals at risk for sustained deterioration of kidney function.


Genome-Wide Association Study , Kidney , AMP-Activated Protein Kinases , Creatinine , Glomerular Filtration Rate/genetics , Humans , Protein Disulfide-Isomerases , United Kingdom
7.
Nat Genet ; 51(10): 1459-1474, 2019 10.
Article En | MEDLINE | ID: mdl-31578528

Elevated serum urate levels cause gout and correlate with cardiometabolic diseases via poorly understood mechanisms. We performed a trans-ancestry genome-wide association study of serum urate in 457,690 individuals, identifying 183 loci (147 previously unknown) that improve the prediction of gout in an independent cohort of 334,880 individuals. Serum urate showed significant genetic correlations with many cardiometabolic traits, with genetic causality analyses supporting a substantial role for pleiotropy. Enrichment analysis, fine-mapping of urate-associated loci and colocalization with gene expression in 47 tissues implicated the kidney and liver as the main target organs and prioritized potentially causal genes and variants, including the transcriptional master regulators in the liver and kidney, HNF1A and HNF4A. Experimental validation showed that HNF4A transactivated the promoter of ABCG2, encoding a major urate transporter, in kidney cells, and that HNF4A p.Thr139Ile is a functional variant. Transcriptional coregulation within and across organs may be a general mechanism underlying the observed pleiotropy between urate and cardiometabolic traits.


Cardiovascular Diseases/blood , Genetic Markers , Gout/blood , Metabolic Diseases/blood , Polymorphism, Single Nucleotide , Signal Transduction , Uric Acid/blood , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Cohort Studies , Genetic Loci , Genetic Predisposition to Disease , Genome-Wide Association Study , Gout/epidemiology , Gout/genetics , Hepatocyte Nuclear Factor 1-alpha/genetics , Hepatocyte Nuclear Factor 4/genetics , Humans , Kidney/metabolism , Kidney/pathology , Liver/metabolism , Liver/pathology , Metabolic Diseases/epidemiology , Metabolic Diseases/genetics , Neoplasm Proteins/genetics , Organ Specificity
8.
Nat Commun ; 10(1): 4130, 2019 09 11.
Article En | MEDLINE | ID: mdl-31511532

Increased levels of the urinary albumin-to-creatinine ratio (UACR) are associated with higher risk of kidney disease progression and cardiovascular events, but underlying mechanisms are incompletely understood. Here, we conduct trans-ethnic (n = 564,257) and European-ancestry specific meta-analyses of genome-wide association studies of UACR, including ancestry- and diabetes-specific analyses, and identify 68 UACR-associated loci. Genetic correlation analyses and risk score associations in an independent electronic medical records database (n = 192,868) reveal connections with proteinuria, hyperlipidemia, gout, and hypertension. Fine-mapping and trans-Omics analyses with gene expression in 47 tissues and plasma protein levels implicate genes potentially operating through differential expression in kidney (including TGFB1, MUC1, PRKCI, and OAF), and allow coupling of UACR associations to altered plasma OAF concentrations. Knockdown of OAF and PRKCI orthologs in Drosophila nephrocytes reduces albumin endocytosis. Silencing fly PRKCI further impairs slit diaphragm formation. These results generate a priority list of genes and pathways for translational research to reduce albuminuria.


Albuminuria/genetics , Chromosome Mapping , Genome-Wide Association Study , Meta-Analysis as Topic , Animals , Creatinine/urine , Diabetes Mellitus/genetics , Diabetes Mellitus/urine , Drosophila melanogaster/genetics , Gene Expression Regulation , Genetic Loci , Genetic Predisposition to Disease , Humans , Phenomics , Risk Factors
9.
Nat Genet ; 51(6): 957-972, 2019 06.
Article En | MEDLINE | ID: mdl-31152163

Chronic kidney disease (CKD) is responsible for a public health burden with multi-systemic complications. Through trans-ancestry meta-analysis of genome-wide association studies of estimated glomerular filtration rate (eGFR) and independent replication (n = 1,046,070), we identified 264 associated loci (166 new). Of these, 147 were likely to be relevant for kidney function on the basis of associations with the alternative kidney function marker blood urea nitrogen (n = 416,178). Pathway and enrichment analyses, including mouse models with renal phenotypes, support the kidney as the main target organ. A genetic risk score for lower eGFR was associated with clinically diagnosed CKD in 452,264 independent individuals. Colocalization analyses of associations with eGFR among 783,978 European-ancestry individuals and gene expression across 46 human tissues, including tubulo-interstitial and glomerular kidney compartments, identified 17 genes differentially expressed in kidney. Fine-mapping highlighted missense driver variants in 11 genes and kidney-specific regulatory variants. These results provide a comprehensive priority list of molecular targets for translational research.


Genetic Association Studies/methods , Genetic Predisposition to Disease , Quantitative Trait Loci , Quantitative Trait, Heritable , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/physiopathology , Chromosome Mapping , Genome-Wide Association Study , Glomerular Filtration Rate , Humans , Inheritance Patterns , Kidney Function Tests , Phenotype , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/urine , Uromodulin/urine , White People
10.
JCI Insight ; 3(23)2018 12 06.
Article En | MEDLINE | ID: mdl-30518689

Biallelic loss-of-function mutations in TRIP11, encoding the golgin GMAP-210, cause the lethal human chondrodysplasia achondrogenesis 1A (ACG1A). We now find that a homozygous splice-site mutation of the lamin B receptor (LBR) gene results in the same phenotype. Intrigued by the genetic heterogeneity, we compared GMAP-210- and LBR-deficient primary cells to unravel how particular mutations in LBR cause a phenocopy of ACG1A. We could exclude a regulatory interaction between LBR and GMAP-210 in patients' cells. However, we discovered a common disruption of Golgi apparatus architecture that was accompanied by decreased secretory trafficking in both cases. Deficiency of Golgi-dependent glycan processing indicated a similar downstream effect of the disease-causing mutations upon Golgi function. Unexpectedly, our results thus point to a common pathogenic mechanism in GMAP-210- and LBR-related diseases attributable to defective secretory trafficking at the Golgi apparatus.


Achondroplasia/genetics , Nuclear Proteins/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Transcription Factors/genetics , Achondroplasia/pathology , Biological Transport, Active/genetics , Cell Proliferation , Cell Survival , Cholesterol/analysis , Cytoskeletal Proteins , Endoplasmic Reticulum/ultrastructure , Female , Fetus , Fibroblasts/pathology , Genetic Diseases, Inborn/genetics , Golgi Apparatus/physiology , Golgi Apparatus/ultrastructure , Humans , Mutation , Pedigree , Phenotype , Sequence Analysis, Protein , Sterols/analysis , Lamin B Receptor
11.
Europace ; 20(12): 2003-2013, 2018 12 01.
Article En | MEDLINE | ID: mdl-29947763

Aims: Characterization of the cardiac phenotype associated with the novel LMNA nonsense mutation c.544C>T, p.Q182*, which we have identified in a large five-generation family. Methods and results: A family tree was constructed. Clinical data [arrhythmia, syncope, sudden cardiac death (SCD), New York Heart Association (NYHA) class] were collected from living and deceased family members. DNA of 23 living family members was analysed for mutations in LMNA. Additionally, dilated cardiomyopathy multi-gene-panel testing and whole exome sequencing were performed in some family members to identify potential phenotype-modifiers. In this five-generation family (n = 65), 17 SCDs occurred at 49.3 ± 10.0 years. Furthermore, we identified eight additional mutation-carriers, seven symptomatic (44 ± 13 years), and one asymptomatic (44 years). First signs of disease [sinus bradycardia with atrioventricular (AV)-block I°] occurred at 36.5 ± 8.1 years. Paroxysmal atrial fibrillation (AF) (onset at 41.8 ± 5.7 years) rapidly progressed to permanent AF (46.2 ± 9.8 years). Subsequently, AV-conduction worsened, syncope, pacemaker-dependence, and non-sustained ventricular tachycardia (43.3 ± 8.2 years) followed. Ventricular arrhythmia caused SCD in patients without implantable cardioverter-defibrillator (ICD). Patients protected by ICD developed rapidly progressive heart failure (45.2 ± 10.6 years). A different phenotype was seen in a sub-family in three patients with early onset of rapidly decompensating heart failure and only minor prior arrhythmia-related symptoms. One patient received high-urgency heart transplantation (HTX) at 32 years, while two died prior to HTX. One of them developed lethal peripartum-associated heart failure. Possible disease-modifiers were identified in this 'heart failure sub-family'. Conclusion: The novel LMNA nonsense mutation c.544C>T causes a severe arrhythmogenic phenotype manifesting with high incidence of SCD in most patients; and in one sub-family, a distinct phenotype with fast progressing heart failure, indicating the need for early consideration of ICD-implantation and listing for heart-transplantation.


Arrhythmias, Cardiac/genetics , Cardiomyopathy, Dilated/genetics , Codon, Nonsense , Death, Sudden, Cardiac/etiology , Lamin Type A/genetics , Adult , Arrhythmias, Cardiac/mortality , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/therapy , Cardiomyopathy, Dilated/mortality , Cardiomyopathy, Dilated/physiopathology , Cardiomyopathy, Dilated/therapy , Death, Sudden, Cardiac/prevention & control , Defibrillators, Implantable , Disease Progression , Electric Countershock/instrumentation , Female , Genetic Predisposition to Disease , Heart Transplantation , Heredity , Humans , Male , Middle Aged , Pedigree , Phenotype , Prognosis , Risk Factors , Severity of Illness Index
12.
PLoS One ; 11(9): e0162466, 2016.
Article En | MEDLINE | ID: mdl-27612175

Genome-wide association studies (GWAS) evaluate associations between genetic variants and a trait or disease of interest free of prior biological hypotheses. GWAS require stringent correction for multiple testing, with genome-wide significance typically defined as association p-value <5*10-8. This study presents a new tool that uses external information about genes to prioritize SNP associations (GenToS). For a given list of candidate genes, GenToS calculates an appropriate statistical significance threshold and then searches for trait-associated variants in summary statistics from human GWAS. It thereby allows for identifying trait-associated genetic variants that do not meet genome-wide significance. The program additionally tests for enrichment of significant candidate gene associations in the human GWAS data compared to the number expected by chance. As proof of principle, this report used external information from a comprehensive resource of genetically manipulated and systematically phenotyped mice. Based on selected murine phenotypes for which human GWAS data for corresponding traits were publicly available, several candidate gene input lists were derived. Using GenToS for the investigation of candidate genes underlying murine skeletal phenotypes in data from a large human discovery GWAS meta-analysis of bone mineral density resulted in the identification of significantly associated variants in 29 genes. Index variants in 28 of these loci were subsequently replicated in an independent GWAS replication step, highlighting that they are true positive associations. One signal, COL11A1, has not been discovered through GWAS so far and represents a novel human candidate gene for altered bone mineral density. The number of observed genes that contained significant SNP associations in human GWAS based on murine candidate gene input lists was much greater than the number expected by chance across several complex human traits (enrichment p-value as low as 10-10). GenToS can be used with any candidate gene list, any GWAS summary file, runs on a desktop computer and is freely available.


Genome-Wide Association Study/methods , Animals , Bone and Bones/metabolism , Collagen Type XI/genetics , Genetic Variation/genetics , Humans , Mice , Phenotype , Polymorphism, Single Nucleotide/genetics
13.
Kidney Int ; 90(4): 764-73, 2016 10.
Article En | MEDLINE | ID: mdl-27263491

Genomewide association studies have identified numerous chronic kidney disease-associated genetic variants, but often do not pinpoint causal genes. This limitation was addressed by combining Mouse Genome Informatics with human genomewide association studies of kidney function. Genes for which mouse models showed abnormal renal physiology, morphology, glomerular filtration rate (GFR), or urinary albumin-to-creatinine ratio were identified from Mouse Genome Informatics. The corresponding human orthologs were then evaluated for GFR-associated single-nucleotide polymorphisms in 133,814 individuals and urinary albumin-to-creatinine ratio-associated SNPs in 54,451 individuals in genome-wide association studies meta-analysis of the CKDGen Consortium. After multiple testing corrections, significant associations with estimated GFR in humans were identified for single-nucleotide polymorphisms in 2, 7, and 17 genes causing abnormal GFR, abnormal physiology, and abnormal morphology in mice, respectively. Genes identified for abnormal kidney morphology showed significant enrichment for estimated GFR-associated single-nucleotide polymorphisms. In total, 19 genes contained variants associated with estimated GFR or the urinary albumin-to-creatinine ratio of which 16 mapped into previously reported genomewide significant loci. CYP26A1 and BMP4 emerged as novel signals subsequently validated in a large, independent study. An additional gene, CYP24A1, was discovered after conditioning on a published nearby association signal. Thus, our novel approach to combine comprehensive mouse phenotype information with human genomewide association studies data resulted in the identification of candidate genes for kidney disease pathogenesis.


Glomerular Filtration Rate , Kidney/physiology , Renal Insufficiency, Chronic/genetics , Albuminuria/genetics , Animals , Bone Morphogenetic Protein 4/genetics , Creatinine/urine , Datasets as Topic , Disease Models, Animal , Genome-Wide Association Study , Humans , Kidney/anatomy & histology , Kidney Function Tests , Mice , Mice, Knockout , Phenotype , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/urine , Retinoic Acid 4-Hydroxylase/genetics , Vitamin D3 24-Hydroxylase/genetics
14.
Nephrol Dial Transplant ; 31(2): 262-9, 2016 Feb.
Article En | MEDLINE | ID: mdl-26420894

BACKGROUND: Chronic kidney disease (CKD) in children is characterized by rapid progression and a high incidence of end-stage renal disease and therefore constitutes an important health problem. While unbiased genetic screens have identified common risk variants influencing renal function and CKD in adults, the presence and identity of such variants in pediatric CKD are unknown. METHODS: The international Pediatric Investigation for Genetic Factors Linked with Renal Progression (PediGFR) Consortium comprises three pediatric CKD cohorts: Chronic Kidney Disease in Children (CKiD), Effect of Strict Blood Pressure Control and ACE Inhibition on the Progression of CRF in Pediatric Patients (ESCAPE) and Cardiovascular Comorbidity in Children with CKD (4C). Clean genotype data from > 10 million genotyped or imputed single-nucleotide polymorphisms (SNPs) were available for 1136 patients with measurements of serum creatinine at study enrollment. Genome-wide association studies were conducted to relate the SNPs to creatinine-based estimated glomerular filtration rate (eGFR crea) and proteinuria (urinary albumin- or protein-to-creatinine ratio ≥ 300 and ≥ 500 mg/g, respectively). In addition, European-ancestry PediGFR patients (cases) were compared with 1347 European-ancestry children without kidney disease (controls) to identify genetic variants associated with the presence of CKD. RESULTS: SNPs with suggestive association P-values < 1 × 10(-5) were identified in 10 regions for eGFR crea, four regions for proteinuria and six regions for CKD including some plausible biological candidates. No SNP was associated at genome-wide significance (P < 5 × 10(-8)). Investigation of the candidate genes for proteinuria in adults from the general population provided support for a region on chromosome 15 near RSL24D1/UNC13C/RAB27A. Conversely, targeted investigation of genes harboring GFR-associated variants in adults from the general population did not reveal significantly associated SNPs in children with CKD. CONCLUSIONS: Our findings suggest that larger collaborative efforts will be needed to draw reliable conclusions about the presence and identity of common variants associated with eGFR, proteinuria and CKD in pediatric populations.


Genome-Wide Association Study/methods , Glomerular Filtration Rate/physiology , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/genetics , Adolescent , Child , Child, Preschool , Disease Progression , Europe/epidemiology , Female , Genetic Loci , Genotype , Humans , Infant , Male , Morbidity/trends , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/physiopathology , Risk Factors
...