51
|
Josipović J, Šimičević L, Dika Ž, Bulj N, Vrsalović M, Jelaković B. UROMODULIN - A LINK BETWEEN SODIUM EXCRETION AND ALTERATION IN CIRCADIAN BLOOD PRESSURE PATTERN IN PREHYPERTENSIVES. Acta Clin Croat 2023; 62:313-322. [PMID: 38549605 PMCID: PMC10969630 DOI: 10.20471/acc.2023.62.02.09] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 07/20/2020] [Indexed: 04/02/2024] Open
Abstract
Although changes in dietary sodium intake alter blood pressure (BP) in salt-sensitive individuals, pathophysiological mechanisms are still unknown. It has been reported that uromodulin is involved in sodium tubular transport, and genome-wide association studies pointed to UMOD gene as one of the most important gene candidates for arterial hypertension. Our aim was to analyze urinary uromodulin, salt intake and BP in 326 young middle-aged subjects (mean age 36±8 years, 49.4% male). In a subgroup of 175 individuals, ambulatory blood pressure monitoring and echocardiogram were performed. Uromodulin was determined by ELISA. According to the JNC-7 criteria, subjects were classified as optimal BP (n=103, men 72%), prehypertension (PHT) (n=143, men 43%) and hypertension (HT) (n= 80, men 38%). There were no differences in age, salt intake, estimated glomerular filtration rate, sodium excretion and uromodulin among BP groups. However, in PHT subjects, uromodulin was positively associated with fractional sodium excretion and negatively with 24-h sodium excretion and diastolic BP dip. These findings point to the effect of uromodulin on sodium reabsorption along the nephron and consequently circadian BP alteration in prehypertensives.
Collapse
Affiliation(s)
- Josipa Josipović
- Department of Internal Medicine, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
- Catholic University of Croatia, School of Medicine, Zagreb, Croatia
| | - Livija Šimičević
- Department of Laboratory Diagnostics, Zagreb University Hospital Center, School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Živka Dika
- Department of Nephrology, Hypertension, Dialysis and Transplantation, Zagreb University Hospital Center, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| | - Nikola Bulj
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Cardiology and Angiology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Mislav Vrsalović
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Cardiology and Angiology, Sestre milosrdnice University Hospital Center, Zagreb, Croatia
| | - Bojan Jelaković
- Department of Nephrology, Hypertension, Dialysis and Transplantation, Zagreb University Hospital Center, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
52
|
Kim S, Koppitch K, Parvez RK, Guo J, Achieng M, Schnell J, Lindström NO, McMahon AP. Comparative single-cell analyses identify shared and divergent features of human and mouse kidney development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.16.540880. [PMID: 37293066 PMCID: PMC10245679 DOI: 10.1101/2023.05.16.540880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Mammalian kidneys maintain fluid homeostasis through the cellular activity of nephrons and the conjoined collecting system. Each epithelial network originates from distinct progenitor cell populations that reciprocally interact during development. To extend our understanding of human and mouse kidney development, we profiled chromatin organization (ATAC-seq) and gene expression (RNA-seq) in developing human and mouse kidneys. Data were analyzed at a species level and then integrated into a common, cross-species multimodal data set. Comparative analysis of cell types and developmental trajectories identified conserved and divergent features of chromatin organization and linked gene activity, revealing species- and cell-type specific regulatory programs. Identification of human-specific enhancer regions linked through GWAS studies to kidney disease highlights the potential of developmental modeling to provide clinical insight.
Collapse
|
53
|
Chen J, Hui Q, Wang Z, Wilson FP, So-Armah K, Freiberg MS, Justice AC, Xu K, Zhao W, Ammous F, Smith JA, Kardia SL, Gwinn M, Marconi VC, Sun YV. Epigenome-Wide Meta-Analysis Reveals Differential DNA Methylation Associated With Estimated Glomerular Filtration Rate Among African American Men With HIV. Kidney Int Rep 2023; 8:1076-1086. [PMID: 37180517 PMCID: PMC10166785 DOI: 10.1016/j.ekir.2023.02.1085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 03/05/2023] Open
Abstract
Introduction People with HIV (PWH) of African ancestry have faster decline of kidney function and faster progression to end-stage renal disease than PWH of European ancestry. DNA methylation have been associated with kidney function in the general population, however, their relationships are unclear for PWH of African ancestry. Methods We performed epigenome-wide association studies (EWAS) of estimated glomerular filtration rate (eGFR) among PWH of African ancestry in 2 subsets of the Veterans Aging Cohort Study cohort (N = 885), followed by a meta-analysis to combine the results. Replication was conducted among independent African American samples without HIV. Results DNA methylation sites cg17944885 near Zinc Finger Family Member 788 (ZNF788) and Zinc Finger Protein 20 (ZNF20), and cg06930757 in SHANK1 were significantly associated with eGFR among PWH of African ancestry (false discovery rate < 0.05). DNA methylation site cg17944885 was also associated with eGFR among different populations including African Americans without HIV. Conclusions Our study attempted to address an important gap in the literature and to understand the role of DNA methylation in renal diseases in PWH of African ancestry. Replication of cg17944885 among different populations suggests there may be a common pathway for renal diseases progression among PWH and people without HIV, and across different ancestral groups. Our results suggest that genes ZNF788/ZNF20 and SHANK1 could be involved in a pathway linking DNA methylation to renal diseases among PWH and are worth further investigation.
Collapse
Affiliation(s)
- Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Zeyuan Wang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Francis P. Wilson
- Department of Medicine, Yale University School of Medicine, Connecticut, USA
| | - Kaku So-Armah
- Boston University School of Medicine, Massachusetts, USA
| | - Matthew S. Freiberg
- Cardiovascular Medicine Division, Vanderbilt University School of Medicine and Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Amy C. Justice
- Connecticut Veteran Health System, West Haven, Connecticut, USA
- Schools of Medicine and Public Health, Yale University, New Haven, Connecticut, USA
| | - Ke Xu
- Connecticut Veteran Health System, West Haven, Connecticut, USA
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, Michigan, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Sharon L.R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, Michigan, USA
| | - Marta Gwinn
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Vincent C. Marconi
- Hubert Department of Global Health, Rollins School of Public Health, Atlanta, Georgia, USA
- Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| | - Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
- Atlanta Veterans Affairs Health Care System, Decatur, Georgia, USA
| |
Collapse
|
54
|
Han SK, McNulty MT, Benway CJ, Wen P, Greenberg A, Onuchic-Whitford AC, Jang D, Flannick J, Burtt NP, Wilson PC, Humphreys BD, Wen X, Han Z, Lee D, Sampson MG. Mapping genomic regulation of kidney disease and traits through high-resolution and interpretable eQTLs. Nat Commun 2023; 14:2229. [PMID: 37076491 PMCID: PMC10115815 DOI: 10.1038/s41467-023-37691-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 03/27/2023] [Indexed: 04/21/2023] Open
Abstract
Expression quantitative trait locus (eQTL) studies illuminate genomic variants that regulate specific genes and contribute to fine-mapped loci discovered via genome-wide association studies (GWAS). Efforts to maximize their accuracy are ongoing. Using 240 glomerular (GLOM) and 311 tubulointerstitial (TUBE) micro-dissected samples from human kidney biopsies, we discovered 5371 GLOM and 9787 TUBE genes with at least one variant significantly associated with expression (eGene) by incorporating kidney single-nucleus open chromatin data and transcription start site distance as an "integrative prior" for Bayesian statistical fine-mapping. The use of an integrative prior resulted in higher resolution eQTLs illustrated by (1) smaller numbers of variants in credible sets with greater confidence, (2) increased enrichment of partitioned heritability for GWAS of two kidney traits, (3) an increased number of variants colocalized with the GWAS loci, and (4) enrichment of computationally predicted functional regulatory variants. A subset of variants and genes were validated experimentally in vitro and using a Drosophila nephrocyte model. More broadly, this study demonstrates that tissue-specific eQTL maps informed by single-nucleus open chromatin data have enhanced utility for diverse downstream analyses.
Collapse
Affiliation(s)
- Seong Kyu Han
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Michelle T McNulty
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Christopher J Benway
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Pei Wen
- Center for Precision Disease Modeling, University of Maryland, School of Medicine, Baltimore, MD, USA
| | - Anya Greenberg
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA
| | - Ana C Onuchic-Whitford
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dongkeun Jang
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Jason Flannick
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
| | - Noël P Burtt
- Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge, MA, USA
| | - Parker C Wilson
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, MO, USA
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St. Louis, St. Louis, MO, USA
- Department of Developmental Biology, Washington University in St. Louis, St. Louis, MO, USA
| | - Xiaoquan Wen
- Department of Biostatistics, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Zhe Han
- Center for Precision Disease Modeling, University of Maryland, School of Medicine, Baltimore, MD, USA.
| | - Dongwon Lee
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA.
- Manton Center for Orphan Disease Research, Boston Children's Hospital, Boston, MA, USA.
| | - Matthew G Sampson
- Division of Pediatric Nephrology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
- Kidney Disease Initiative, Broad Institute, Cambridge, MA, USA.
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
55
|
Chhabra R, Guergues J, Wohlfahrt J, Rockfield S, Espinoza Gonzalez P, Rego S, Park MA, Berglund AE, Stevens SM, Nanjundan M. Deregulated expression of the 14q32 miRNA cluster in clear cell renal cancer cells. Front Oncol 2023; 13:1048419. [PMID: 37139155 PMCID: PMC10150008 DOI: 10.3389/fonc.2023.1048419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/21/2023] [Indexed: 05/05/2023] Open
Abstract
Clear cell renal cell carcinomas (ccRCC) are characterized by arm-wide chromosomal alterations. Loss at 14q is associated with disease aggressiveness in ccRCC, which responds poorly to chemotherapeutics. The 14q locus contains one of the largest miRNA clusters in the human genome; however, little is known about the contribution of these miRNAs to ccRCC pathogenesis. In this regard, we investigated the expression pattern of selected miRNAs at the 14q32 locus in TCGA kidney tumors and in ccRCC cell lines. We demonstrated that the miRNA cluster is downregulated in ccRCC (and cell lines) as well as in papillary kidney tumors relative to normal kidney tissues (and primary renal proximal tubule epithelial (RPTEC) cells). We demonstrated that agents modulating expression of DNMT1 (e.g., 5-Aza-deoxycytidine) could modulate 14q32 miRNA expression in ccRCC cell lines. Lysophosphatidic acid (LPA, a lysophospholipid mediator elevated in ccRCC) not only increased labile iron content but also modulated expression of a 14q32 miRNA. Through an overexpression approach targeting a subset of 14q32 miRNAs (specifically at subcluster A: miR-431-5p, miR-432-5p, miR-127-3p, and miR-433-3p) in 769-P cells, we uncovered changes in cellular viability and claudin-1, a tight junction marker. A global proteomic approach was implemented using these miRNA overexpressing cell lines which uncovered ATXN2 as a highly downregulated target. Collectively, these findings support a contribution of miRNAs at 14q32 in ccRCC pathogenesis.
Collapse
Affiliation(s)
- Ravneet Chhabra
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - Jennifer Guergues
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - Jessica Wohlfahrt
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - Stephanie Rockfield
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
- Department of Cell and Molecular Biology, St. Jude Children’s Research Hospital, Memphis, TN, United States
| | - Pamela Espinoza Gonzalez
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - Shanon Rego
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - Margaret A. Park
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Anders E. Berglund
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, United States
| | - Stanley M. Stevens
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| | - Meera Nanjundan
- Department of Cell Biology, Microbiology, and Molecular Biology, University of South Florida, Tampa, FL, United States
| |
Collapse
|
56
|
Wagner CA, Unwin R, Lopez-Garcia SC, Kleta R, Bockenhauer D, Walsh S. The pathophysiology of distal renal tubular acidosis. Nat Rev Nephrol 2023; 19:384-400. [PMID: 37016093 DOI: 10.1038/s41581-023-00699-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
The kidneys have a central role in the control of acid-base homeostasis owing to bicarbonate reabsorption and production of ammonia and ammonium in the proximal tubule and active acid secretion along the collecting duct. Impaired acid excretion by the collecting duct system causes distal renal tubular acidosis (dRTA), which is characterized by the failure to acidify urine below pH 5.5. This defect originates from reduced function of acid-secretory type A intercalated cells. Inherited forms of dRTA are caused by variants in SLC4A1, ATP6V1B1, ATP6V0A4, FOXI1, WDR72 and probably in other genes that are yet to be discovered. Inheritance of dRTA follows autosomal-dominant and -recessive patterns. Acquired forms of dRTA are caused by various types of autoimmune diseases or adverse effects of some drugs. Incomplete dRTA is frequently found in patients with and without kidney stone disease. These patients fail to appropriately acidify their urine when challenged, suggesting that incomplete dRTA may represent an intermediate state in the spectrum of the ability to excrete acids. Unrecognized or insufficiently treated dRTA can cause rickets and failure to thrive in children, osteomalacia in adults, nephrolithiasis and nephrocalcinosis. Electrolyte disorders are also often present and poorly controlled dRTA can increase the risk of developing chronic kidney disease.
Collapse
Affiliation(s)
- Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland.
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK.
| | - Robert Unwin
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Sergio C Lopez-Garcia
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Robert Kleta
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| | - Detlef Bockenhauer
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
- Department of Paediatric Nephrology, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Stephen Walsh
- Department of Renal Medicine, Royal Free Hospital, University College London, London, UK
| |
Collapse
|
57
|
Goto S, Hosojima M, Kabasawa H, Saito A. The endocytosis receptor megalin: From bench to bedside. Int J Biochem Cell Biol 2023; 157:106393. [PMID: 36863658 DOI: 10.1016/j.biocel.2023.106393] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
The large (∼600 kDa) endocytosis receptor megalin/low-density lipoprotein receptor-related protein 2 is highly expressed at the apical membrane of proximal tubular epithelial cells (PTECs). Megalin plays an important role in the endocytosis of various ligands via interactions with intracellular adaptor proteins, which mediate the trafficking of megalin in PTECs. Megalin mediates the retrieval of essential substances, including carrier-bound vitamins and elements, and impairment of the endocytic process may result in the loss of those substances. In addition, megalin reabsorbs nephrotoxic substances such as antimicrobial (colistin, vancomycin, and gentamicin) or anticancer (cisplatin) drugs and advanced glycation end product-modified or fatty acid-containing albumin. The megalin-mediated uptake of these nephrotoxic ligands causes metabolic overload in PTECs and leads to kidney injury. Blockade or suppression of the megalin-mediated endocytosis of nephrotoxic substances may represent a novel therapeutic strategy for drug-induced nephrotoxicity or metabolic kidney disease. Megalin reabsorbs urinary biomarker proteins such as albumin, α1-microglobulin, β2-microglobulin, and liver-type fatty acid-binding protein; thus, the above-mentioned megalin-targeted therapy may have an effect on the urinary excretion of these biomarkers. We have previously established a sandwich enzyme-linked immunosorbent assay to measure the ectodomain (A-megalin) and full-length (C-megalin) forms of urinary megalin using monoclonal antibodies against the amino- and carboxyl-terminals of megalin, respectively, and reported their clinical usefulness. In addition, there have been reports of patients with novel pathological anti-brush border autoantibodies targeting megalin in the kidney. Even with these breakthroughs in the characterization of megalin, a large number of issues remain to be addressed in future research.
Collapse
Affiliation(s)
- Sawako Goto
- Departments of Applied Molecular Medicine, Japan
| | - Michihiro Hosojima
- Departments of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | - Hideyuki Kabasawa
- Departments of Clinical Nutrition Science, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata City, Niigata, Japan
| | | |
Collapse
|
58
|
Kim JY, Chun SY, Lim H, Chang TI. Association between familial aggregation of chronic kidney disease and its incidence and progression. Sci Rep 2023; 13:5131. [PMID: 36991140 PMCID: PMC10060248 DOI: 10.1038/s41598-023-32362-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
This study aimed to examine the association between familial aggregation of chronic kidney disease (CKD) and risk of CKD development and its progression. This nationwide family study comprised 881,453 cases with newly diagnosed CKD between 2004 and 2017 and 881,453 controls without CKD matched by age and sex, using data from the Korean National Health Insurance Service with linkage to the family tree database. The risks of CKD development and disease progression, defined as an incident end-stage renal disease (ESRD), were evaluated. The presence of any affected family member with CKD was associated with a significantly higher risk of CKD with adjusted ORs (95% CI) of 1.42 (1.38-1.45), 1.50 (1.46-1.55), 1.70 (1.64-1.77), and 1.30 (1.27-1.33) for individuals with affected parents, offspring, siblings, and spouses, respectively. In Cox models conducted on patients with predialysis CKD, risk of incident ESRD was significantly higher in those with affected family members with ESRD. The corresponding HRs (95% CI) were 1.10 (1.05-1.15), 1.38 (1.32-1.46), 1.57 (1.49-1.65), and 1.14 (1.08-1.19) for individuals listed above, respectively. Familial aggregation of CKD was strongly associated with a higher risk of CKD development and disease progression to ESRD.
Collapse
Affiliation(s)
- Jae Young Kim
- Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, 100 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10444, Republic of Korea
- Department of Internal Medicine, Institute of Kidney Disease Research, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung-Youn Chun
- Research and Analysis Team, National Health Insurance Service Ilsan Hospital, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Hyunsun Lim
- Research and Analysis Team, National Health Insurance Service Ilsan Hospital, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Tae Ik Chang
- Department of Internal Medicine, National Health Insurance Service Ilsan Hospital, 100 Ilsan-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10444, Republic of Korea.
| |
Collapse
|
59
|
Peng Y, Li Y, Zhang W, ShangGuan Y, Xie T, Wang K, Qiu J, Pu W, Hu B, Zhang X, Yin L, Tang D, Dai Y. The characteristics of extrachromosomal circular DNA in patients with end-stage renal disease. Eur J Med Res 2023; 28:134. [PMID: 36967395 PMCID: PMC10041755 DOI: 10.1186/s40001-023-01064-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 02/15/2023] [Indexed: 03/28/2023] Open
Abstract
BACKGROUND End-stage renal disease (ESRD) is the final stage of chronic kidney disease (CKD). In addition to the structurally intact chromosome genomic DNA, there is a double-stranded circular DNA called extrachromosomal circular DNA (eccDNA), which is thought to be involved in the epigenetic regulation of human disease. However, the features of eccDNA in ESRD patients are barely known. In this study, we identified eccDNA from ESRD patients and healthy people, as well as revealed the characteristics of eccDNA in patients with ESRD. METHODS Using the high-throughput Circle-Sequencing technique, we examined the eccDNA in peripheral blood mononuclear cells (PBMCs) from healthy people (NC) (n = 12) and ESRD patients (n = 16). We analyzed the length distribution, genome elements, and motifs feature of eccDNA in ESRD patients. Then, after identifying the specific eccDNA in ESRD patients, we explored the potential functions of the target genes of the specific eccDNA. Finally, we investigated the probable hub eccDNA using algorithms. RESULTS In total, 14,431 and 11,324 eccDNAs were found in the ESRD and NC groups, respectively, with sizes ranging from 0.01 kb to 60 kb at most. Additionally, the ESRD group had a greater distribution of eccDNA on chromosomes 4, 11, 13, and 20. In two groups, we also discovered several motifs of specific eccDNAs. Furthermore, we identified 13,715 specific eccDNAs in the ESRD group and 10,585 specific eccDNAs in the NC group, both of which were largely annotated as mRNA catalog. Pathway studies using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) showed that the specific eccDNA in ESRD was markedly enriched in cell junction and communication pathways. Furthermore, we identified potentially 20 hub eccDNA-targeting genes from all ESRD-specific eccDNA-targeting genes. Also, we found that 39 eccDNA-targeting genes were associated with ESRD, and some of these eccDNAs may be related to the pathogenesis of ESRD. CONCLUSIONS Our findings revealed the characteristics of eccDNA in ESRD patients and discovered potentially hub and ESRD-relevant eccDNA-targeting genes, suggesting a novel probable mechanism of ESRD.
Collapse
Affiliation(s)
- Yue Peng
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Yixi Li
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Wei Zhang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Yu ShangGuan
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Ting Xie
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Kang Wang
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Jing Qiu
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Wenjun Pu
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China
| | - Biying Hu
- Institute of Nephrology and Blood Purification, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xinzhou Zhang
- Key Renal Laboratory of Shenzhen, Department of Nephrology, Shenzhen People's Hospital, The Second Clinical Medical College of Jinan University, Shenzhen, 518020, Guangdong, China
| | - Lianghong Yin
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China.
- Guangzhou Enttxs Medical Products Co., Ltd. P.R. Guangzhou, Guangzhou, China.
| | - Donge Tang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China.
| | - Yong Dai
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Jinan University, Shenzhen, China.
- Department of Pathology, The 924th Hospital of the Chinese People's Liberation Army Joint Logistic Support Force, Guangxi Key Laboratory of Metabolic Diseases Research, Guilin, 541002, Guangxi, China.
| |
Collapse
|
60
|
Wuttke M, König E, Katsara MA, Kirsten H, Farahani SK, Teumer A, Li Y, Lang M, Göcmen B, Pattaro C, Günzel D, Köttgen A, Fuchsberger C. Imputation-powered whole-exome analysis identifies genes associated with kidney function and disease in the UK Biobank. Nat Commun 2023; 14:1287. [PMID: 36890159 PMCID: PMC9995463 DOI: 10.1038/s41467-023-36864-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 02/20/2023] [Indexed: 03/10/2023] Open
Abstract
Genome-wide association studies have discovered hundreds of associations between common genotypes and kidney function but cannot comprehensively investigate rare coding variants. Here, we apply a genotype imputation approach to whole exome sequencing data from the UK Biobank to increase sample size from 166,891 to 408,511. We detect 158 rare variants and 105 genes significantly associated with one or more of five kidney function traits, including genes not previously linked to kidney disease in humans. The imputation-powered findings derive support from clinical record-based kidney disease information, such as for a previously unreported splice allele in PKD2, and from functional studies of a previously unreported frameshift allele in CLDN10. This cost-efficient approach boosts statistical power to detect and characterize both known and novel disease susceptibility variants and genes, can be generalized to larger future studies, and generates a comprehensive resource ( https://ckdgen-ukbb.gm.eurac.edu/ ) to direct experimental and clinical studies of kidney disease.
Collapse
Affiliation(s)
- Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
- Renal Division, Department of Medicine, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany.
| | - Eva König
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Maria-Alexandra Katsara
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, University of Leipzig, Leipzig, Germany
- LIFE Research Centre for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | | | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Martin Lang
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Burulca Göcmen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Cristian Pattaro
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy
| | - Dorothee Günzel
- Clinical Physiology/Nutritional Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christian Fuchsberger
- Eurac Research, Institute for Biomedicine (affiliated to the University of Lübeck), Bolzano, Italy.
| |
Collapse
|
61
|
Kim YJ, Zhang H, Lee Y, Seymen F, Koruyucu M, Kasimoglu Y, Simmer JP, Hu JCC, Kim JW. Novel WDR72 Mutations Causing Hypomaturation Amelogenesis Imperfecta. J Pers Med 2023; 13:326. [PMID: 36836560 PMCID: PMC9965932 DOI: 10.3390/jpm13020326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023] Open
Abstract
Amelogenesis imperfecta (AI) is a heterogeneous collection of hereditary enamel defects. The affected enamel can be classified as hypoplastic, hypomaturation, or hypocalcified in form. A better understanding of normal amelogenesis and improvements in our ability to diagnose AI through genetic testing can be realized through more complete knowledge of the genes and disease-causing variants that cause AI. In this study, mutational analysis was performed with whole exome sequencing (WES) to identify genetic etiology underlying the hypomaturation AI condition in affected families. Mutational analyses identified biallelic WDR72 mutations in four hypomaturation AI families. Novel mutations include a homozygous deletion and insertion mutation (NM_182758.4: c.2680_2699delinsACTATAGTT, p.(Ser894Thrfs*15)), compound heterozygous mutations (paternal c.2332dupA, p.(Met778Asnfs*4)) and (maternal c.1287_1289del, p.(Ile430del)) and a homozygous 3694 bp deletion that includes exon 14 (NG_017034.2:g.96472_100165del). A homozygous recurrent mutation variant (c.1467_1468delAT, p.(Val491Aspfs*8)) was also identified. Current ideas on WDR72 structure and function are discussed. These cases expand the mutational spectrum of WDR72 mutations causing hypomaturation AI and improve the possibility of genetic testing to accurately diagnose AI caused by WDR72 defects.
Collapse
Affiliation(s)
- Youn Jung Kim
- Department of Pediatric Dentistry & DRI, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Hong Zhang
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yejin Lee
- Department of Pediatric Dentistry & DRI, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Figen Seymen
- Department of Paediatric Dentistry, Faculty of Dentistry, Altinbas University, Istanbul 34147, Turkey
| | - Mine Koruyucu
- Department of Pedodontics, Faculty of Dentistry, Istanbul University, Istanbul 34116, Turkey
| | - Yelda Kasimoglu
- Department of Pedodontics, Faculty of Dentistry, Istanbul University, Istanbul 34116, Turkey
| | - James P. Simmer
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jan C.-C. Hu
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jung-Wook Kim
- Department of Pediatric Dentistry & DRI, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
- Department of Molecular Genetics & DRI, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| |
Collapse
|
62
|
Zhu S, Li W, Zhang H, Yan Y, Mei Q, Wu K. Retinal determination gene networks: from biological functions to therapeutic strategies. Biomark Res 2023; 11:18. [PMID: 36750914 PMCID: PMC9906957 DOI: 10.1186/s40364-023-00459-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/28/2023] [Indexed: 02/09/2023] Open
Abstract
The retinal determinant gene network (RDGN), originally discovered as a critical determinator in Drosophila eye specification, has become an important regulatory network in tumorigenesis and progression, as well as organogenesis. This network is not only associated with malignant biological behaviors of tumors, such as proliferation, and invasion, but also regulates the development of multiple mammalian organs. Three members of this conservative network have been extensively investigated, including DACH, SIX, and EYA. Dysregulated RDGN signaling is associated with the initiation and progression of tumors. In recent years, it has been found that the members of this network can be used as prognostic markers for cancer patients. Moreover, they are considered to be potential therapeutic targets for cancer. Here, we summarize the research progress of RDGN members from biological functions to signaling transduction, especially emphasizing their effects on tumors. Additionally, we discuss the roles of RDGN members in the development of organs and tissue as well as their correlations with the pathogenesis of chronic kidney disease and coronary heart disease. By summarizing the roles of RDGN members in human diseases, we hope to promote future investigations into RDGN and provide potential therapeutic strategies for patients.
Collapse
Affiliation(s)
- Shuangli Zhu
- grid.412793.a0000 0004 1799 5032Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wanling Li
- grid.412793.a0000 0004 1799 5032Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,grid.470966.aCancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032 China
| | - Hao Zhang
- grid.412793.a0000 0004 1799 5032Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yuheng Yan
- grid.412793.a0000 0004 1799 5032Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. .,Cancer Center, Tongji hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
63
|
The genetic side of diabetic kidney disease: a review. Int Urol Nephrol 2023; 55:335-343. [PMID: 35974289 DOI: 10.1007/s11255-022-03319-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/24/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Diabetic kidney disease (DKD) is one of the most common complications of diabetes, with approximately 30-40% of patients with type 1 diabetes mellitus and 20% of patients with type 2 diabetes mellitus eventually developing DKD. If DKD is not controlled in the early clinical stage and proteinuria develops, the disease will progress to end-stage renal disease. The pathogenesis of DKD remains largely unknown and is multifactorial, likely due to interactions between genetic and environmental factors. Familial clustering also supports a critical role of hereditary factors in DKD. The development of gene detection technology has promoted the exploration of DKD susceptibility genes in different cohorts of patients with diabetes. Identifying susceptibility genes can provide insights into the pathogenesis of DKD, as well as a basis for its clinical diagnosis and therapy. RESULTS Numerous candidate gene loci have been found to be associated with DKD, many of which play critical regulatory roles in the pathogenesis of this disease, including genes involved in glycol-metabolism, lipid metabolism, the renin-angiotensin-aldosterone system, inflammation and oxidative stress. In this review, we summarize the functions of several susceptibility genes involved in the development of DKD. CONCLUSION Based on our findings, we recommend that studying susceptibility gene polymorphisms can lead to a better understanding of the pathogenesis of DKD and could help prevent this disease or improve its outcomes.
Collapse
|
64
|
Durand A, Winkler CA, Vince N, Douillard V, Geffard E, Binns-Roemer E, Ng DK, Gourraud PA, Reidy K, Warady B, Furth S, Kopp JB, Kaskel FJ, Limou S. Identification of Novel Genetic Risk Factors for Focal Segmental Glomerulosclerosis in Children: Results From the Chronic Kidney Disease in Children (CKiD) Cohort. Am J Kidney Dis 2023; 81:635-646.e1. [PMID: 36623684 DOI: 10.1053/j.ajkd.2022.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 11/02/2022] [Indexed: 01/09/2023]
Abstract
RATIONALE & OBJECTIVE Focal segmental glomerulosclerosis (FSGS) is a major cause of pediatric nephrotic syndrome, and African Americans exhibit an increased risk for developing FSGS compared with other populations. Predisposing genetic factors have previously been described in adults. Here we performed genomic screening of primary FSGS in a pediatric African American population. STUDY DESIGN Prospective cohort with case-control genetic association study design. SETTING & PARTICIPANTS 140 African American children with chronic kidney disease from the Chronic Kidney Disease in Children (CKiD) cohort, including 32 cases with FSGS. PREDICTORS Over 680,000 common single-nucleotide polymorphisms (SNPs) were tested for association. We also ran a pathway enrichment analysis and a human leucocyte antigen (HLA)-focused association study. OUTCOME Primary biopsy-proven pediatric FSGS. ANALYTICAL APPROACH Multivariate logistic regression models. RESULTS The genome-wide association study revealed 169 SNPs from 14 independent loci significantly associated with FSGS (false discovery rate [FDR]<5%). We observed notable signals for genetic variants within the APOL1 (P=8.6×10-7; OR, 25.8 [95% CI, 7.1-94.0]), ALMS1 (P=1.3×10-7; 13.0% in FSGS cases vs 0% in controls), and FGFR4 (P=4.3×10-6; OR, 24.8 [95% CI, 6.3-97.7]) genes, all of which had previously been associated with adult FSGS, kidney function, or chronic kidney disease. We also highlighted novel, functionally relevant genes, including GRB2 (which encodes a slit diaphragm protein promoting podocyte structure through actin polymerization) and ITGB1 (which is linked to renal injuries). Our results suggest a major role for immune responses and antigen presentation in pediatric FSGS through (1) associations with SNPs in PTPRJ (or CD148, P=3.5×10-7), which plays a role in T-cell receptor signaling, (2) HLA-DRB1∗11:01 association (P=6.1×10-3; OR, 4.5 [95% CI, 1.5-13.0]), and (3) signaling pathway enrichment (P=1.3×10-6). LIMITATIONS Sample size and no independent replication cohort with genomic data readily available. CONCLUSIONS Our genetic study has identified functionally relevant risk factors and the importance of immune regulation for pediatric primary FSGS, which contributes to a better description of its molecular pathophysiological mechanisms.
Collapse
Affiliation(s)
- Axelle Durand
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Cheryl A Winkler
- Basic Research Laboratory, Center for Cancer Research, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland
| | - Nicolas Vince
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Venceslas Douillard
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Estelle Geffard
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Elizabeth Binns-Roemer
- Basic Research Laboratory, Center for Cancer Research, Frederick National Laboratory, National Cancer Institute, Frederick, Maryland
| | - Derek K Ng
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Pierre-Antoine Gourraud
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France
| | - Kimberley Reidy
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | | | - Susan Furth
- Children's Hospital of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey B Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland
| | - Frederick J Kaskel
- Children's Hospital at Montefiore, Albert Einstein College of Medicine, Bronx, New York
| | - Sophie Limou
- Center for Research in Transplantation and Translational Immunology (UMR 1064), Nantes Université, Ecole Centrale Nantes, CHU Nantes, INSERM, F-44000 Nantes, France.
| |
Collapse
|
65
|
Liu Y, Chen Y, Yang Q, Shen D, Du Z, Zhang G. Single nucleotide polymorphisms in the GFR-related gene and the SNP-SNP interactions on the risk of diabetic kidney disease in Chinese Han population. Acta Diabetol 2023; 60:115-125. [PMID: 36378321 DOI: 10.1007/s00592-022-01988-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 09/29/2022] [Indexed: 11/16/2022]
Abstract
PURPOSE Genetic susceptibility is an important pathogenic mechanism in diabetic kidney disease (DKD). However, the specific gene variant associated with DKD susceptibility remains unclear. Glomerular filtration rate (GFR), an important indicator for the process of DKD, has a heritable component. This study aimed to explore whether these GFR-related single nucleotide polymorphisms (SNPs) were associated with DKD. METHODS GFR-related SNPs were collected from the Phenotype-Genotype Integrator (PheGenI) database. SNPs for population cohort analysis were selected following the criteria of complete records of eQTL and MAF > 5% in the Chinese Han population. Totally 498 subjects participated, including166 patients with DKD, 166 patients with T2DM, and 166 controls. The genotypes of SNPs were determined using a Sequenom MassARRAY system. Plink software was employed to analyze the SNP-SNP interactions. RESULTS By screening the GFR-related SNPs recorded in the PheGenI database, four SNPs (rs1260326, rs17319721, rs35716097, and rs6420094) were finally selected to investigate the association with DKD. It was shown that one of the four SNPs was related to DKD. The G allele of SLC34A1 rs6420094 was associated with a decreased risk of DKD in DKD and T2DM groups (OR 0.716; P = 0.049). Genetic model analysis revealed that rs6420094 was a protective factor for DKD in T2DM in a dominant model and an additive model (P = 0.03; P = 0.032, respectively). Although rs17319721 was not associated with the risk of DKD, the SNP-SNP interactions between rs17319721 and rs6420094 predicted a significantly decreased risk of DKD (OR 0.464; P = 0.047). CONCLUSION SLC34A1 rs6420094 was associated with a decreased DKD risk in the Chinese Han population. SNP-SNP interaction between rs17319721 and rs6420094 was associated with a lower risk of DKD.
Collapse
Affiliation(s)
- Yanxiu Liu
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China
- Department of Pediatric Nephrology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yan Chen
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Qiwei Yang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Dihan Shen
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zhenwu Du
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China.
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China.
| | - Guizhen Zhang
- Medical Research Center, The Second Hospital of Jilin University, Changchun, Jilin, China.
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
66
|
Wruck W, Boima V, Erichsen L, Thimm C, Koranteng T, Kwakyi E, Antwi S, Adu D, Adjaye J. Urine-Based Detection of Biomarkers Indicative of Chronic Kidney Disease in a Patient Cohort from Ghana. J Pers Med 2022; 13:jpm13010038. [PMID: 36675700 PMCID: PMC9863148 DOI: 10.3390/jpm13010038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic kidney disease (CKD) is a global health burden with a continuously increasing prevalence associated with an increasing incidence of diabetes and hypertension in aging populations. CKD is characterized by low glomerular filtration rate (GFR) and other renal impairments including proteinuria, thus implying that multiple factors may contribute to the etiology this disease. While there are indications of ethnic differences, it is hard to disentangle these from confounding social factors. Usually, CKD is detected in later stages of the disease when irreversible renal damage has already occurred, thus suggesting a need for early non-invasive diagnostic markers. In this study, we explored the urine secretome of a CKD patient cohort from Ghana with 40 gender-matched patients and 40 gender-matched healthy controls employing a kidney injury and a more general cytokine assay. We identified panels of kidney-specific cytokine markers, which were also gender-specific, and a panel of gender-independent cytokine markers. The gender-specific markers are IL10 and MME for male and CLU, RETN, AGER, EGFR and VEGFA for female. The gender-independent cytokine markers were APOA1, ANGPT2, C5, CFD, GH1, ICAM1, IGFBP2, IL8, KLK4, MMP9 and SPP1 (up-regulated) and FLT3LG, CSF1, PDGFA, RETN and VEGFA (down-regulated). APOA1-the major component of HDL particles-was up-regulated in Ghanaian CKD patients and its co-occurrence with APOL1 in a subpopulation of HDL particles may point to specific CKD-predisposing APOL1 haplotypes in patients of African descent-this, however, needs further investigation. The identified panels, though preliminary, lay down the foundation for the development of robust CKD-diagnostic assays.
Collapse
Affiliation(s)
- Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Vincent Boima
- Department of Medicine & Therapeutics, University of Ghana Medical School, College of Health Sciences, Box 4236, University of Ghana, Accra P.O. Box LG 1181, Ghana
| | - Lars Erichsen
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Chantelle Thimm
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
| | - Theresa Koranteng
- NHS-Clover Health Centre, Equitable House, 10 Woolich New Road, Woolich, London SE18 6AB, UK
| | - Edward Kwakyi
- Department of Medicine & Therapeutics, University of Ghana Medical School, College of Health Sciences, Box 4236, University of Ghana, Accra P.O. Box LG 1181, Ghana
| | - Sampson Antwi
- Department of Child Health, School of Medical Sciences, Kwame Nkrumah University of Science and Technology, Komfo Anokye Teaching Hospital, Kumasi P.O. Box KS 9265, Ghana
| | - Dwomoa Adu
- Department of Medicine & Therapeutics, University of Ghana Medical School, College of Health Sciences, Box 4236, University of Ghana, Accra P.O. Box LG 1181, Ghana
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstr. 5, 40225 Düsseldorf, Germany
- EGA Institute for Women’s Health, University College London, 86-96 Chenies Mews, London WC1E 6HX, UK
- Correspondence:
| |
Collapse
|
67
|
Ruilope LM, Ortiz A, Lucia A, Miranda B, Alvarez-Llamas G, Barderas MG, Volpe M, Ruiz-Hurtado G, Pitt B. Prevention of cardiorenal damage: importance of albuminuria. Eur Heart J 2022; 44:1112-1123. [PMID: 36477861 DOI: 10.1093/eurheartj/ehac683] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 10/20/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Abstract
Chronic kidney disease (CKD) is projected to become a leading global cause of death by 2040, and its early detection is critical for effective and timely management. The current definition of CKD identifies only advanced stages, when kidney injury has already destroyed >50% of functioning kidney mass as reflected by an estimated glomerular filtration rate <60 mL/min/1.73 m2 or a urinary albumin/creatinine ratio >six-fold higher than physiological levels (i.e. > 30 mg/g). An elevated urinary albumin-excretion rate is a known early predictor of future cardiovascular events. There is thus a ‘blind spot’ in the detection of CKD, when kidney injury is present but is undetectable by current diagnostic criteria, and no intervention is made before renal and cardiovascular damage occurs. The present review discusses the CKD ‘blind spot’ concept and how it may facilitate a holistic approach to CKD and cardiovascular disease prevention and implement the call for albuminuria screening implicit in current guidelines. Cardiorenal risk associated with albuminuria in the high-normal range, novel genetic and biochemical markers of elevated cardiorenal risk, and the role of heart and kidney protective drugs evaluated in recent clinical trials are also discussed. As albuminuria is a major risk factor for cardiovascular and renal disease, starting from levels not yet considered in the definition of CKD, the implementation of opportunistic or systematic albuminuria screening and therapy, possibly complemented with novel early biomarkers, has the potential to improve cardiorenal outcomes and mitigate the dismal 2040 projections for CKD and related cardiovascular burden.
Collapse
Affiliation(s)
- Luis M Ruilope
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario , 12 de Octubre, Avenida de Córdoba s/n , Spain
- CIBER-CV, Hospital Universitario , Av. de Córdoba s/n, 28041, Madrid , Spain
- Faculty of Sport Sciences, Universidad Europea de Madrid , Tajo, s/n, 28670 Villaviciosa de Odón, Madrid , Spain
| | - Alberto Ortiz
- IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Av. de los Reyes Católicos, 2, 28040 Madrid , Spain
- RICORS2040, Hospital Universitario Fundación Jiménez Díaz , Madrid , Spain
| | - Alejandro Lucia
- Faculty of Sport Sciences, Universidad Europea de Madrid , Tajo, s/n, 28670 Villaviciosa de Odón, Madrid , Spain
| | - Blanca Miranda
- Fundación Renal Íñigo Álvarez de Toledo , José Abascal, 42, 28003 Madrid , Spain
| | - Gloria Alvarez-Llamas
- IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid , Av. de los Reyes Católicos, 2, 28040 Madrid , Spain
- RICORS2040, Hospital Universitario Fundación Jiménez Díaz , Madrid , Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos (HNP), SESCAM , FINCA DE, Carr. de la Peraleda, S/N, 45004 Toledo , Spain
| | - Massimo Volpe
- Cardiology, Department of Clinical and Molecular Medicine, Sapienza University of Rome and IRCCS San Raffaele Rome , Sant'Andrea Hospital, Rome , Italy
| | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Institute of Research Imas12, Hospital Universitario , 12 de Octubre, Avenida de Córdoba s/n , Spain
- CIBER-CV, Hospital Universitario , Av. de Córdoba s/n, 28041, Madrid , Spain
| | - Bertram Pitt
- Division of Cardiology, University of Michigan School of Medicine , Ann Arbor, MI , USA
| |
Collapse
|
68
|
Wang K, Shi M, Yang A, Fan B, Tam CHT, Lau E, Luk AOY, Kong APS, Ma RCW, Chan JCN, Chow E. GCKR and GCK polymorphisms are associated with increased risk of end-stage kidney disease in Chinese patients with type 2 diabetes: The Hong Kong Diabetes Register (1995-2019). Diabetes Res Clin Pract 2022; 193:110118. [PMID: 36243233 DOI: 10.1016/j.diabres.2022.110118] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/30/2022] [Accepted: 10/06/2022] [Indexed: 11/26/2022]
Abstract
AIMS Glucokinase (GCK) and glucokinase regulatory protein (GKRP) regulate glucose and lipid metabolism. We investigated the associations of GCKR and GCK polymorphisms with kidney outcomes. METHODS Analyses were performed in a prospective cohort who were enrolled in the Hong Kong Diabetes Register between 1995 and 2017. The associations of GCKR rs1260326 and GCK rs1799884 polymorphisms with incident end-stage kidney disease (ESKD), albuminuria and rapid eGFR decline were analysed by Cox regression or logistic regression with adjustment. RESULTS 6072 patients (baseline mean age 57.4 years; median diabetes duration 6.0 years; 54.5 % female) were included, with a median follow-up of 15.5 years. The GCKR rs1260326 [HR (95 %CI) 1.23 (1.05-1.44) for CT; HR 1.23 (1.02-1.48) for TT] and GCK rs1799884 T alleles [HR 1.73 (1.24-2.40) for TT] were independently associated with increased risk of ESKD versus their respective CC genotypes. GCKR rs1260326 T allele was also associated with albuminuria [OR 1.18 (1.05-1.33) for CT; OR 1.34 (1.16-1.55) for TT] and rapid eGFR decline. CONCLUSIONS In Chinese patients with type 2 diabetes, T allele carriers of GCKR rs1260326 and GCK rs1799884 were at high risk for ESKD. These genetic markers may be used to identify high risk patients for early intensive management for renoprotection.
Collapse
Affiliation(s)
- Ke Wang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Mai Shi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Aimin Yang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Baoqi Fan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Claudia H T Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Eric Lau
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Andrea O Y Luk
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Alice P S Kong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China.
| | - Elaine Chow
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China; Phase 1 Clinical Trial Centre, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong Special Administrative Region, China.
| |
Collapse
|
69
|
Mary S, Boder P, Padmanabhan S, McBride MW, Graham D, Delles C, Dominiczak AF. Role of Uromodulin in Salt-Sensitive Hypertension. Hypertension 2022; 79:2419-2429. [PMID: 36378920 PMCID: PMC9553220 DOI: 10.1161/hypertensionaha.122.19888] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The exclusive expression of uromodulin in the kidneys has made it an intriguing protein in kidney and cardiovascular research. Genome-wide association studies discovered variants of uromodulin that are associated with chronic kidney diseases and hypertension. Urinary and circulating uromodulin levels reflect kidney and cardiovascular health as well as overall mortality. More recently, Mendelian randomization studies have shown that genetically driven levels of uromodulin have a causal and adverse effect on kidney function. On a mechanistic level, salt sensitivity is an important factor in the pathophysiology of hypertension, and uromodulin is involved in salt reabsorption via the NKCC2 (Na+-K+-2Cl- cotransporter) on epithelial cells of the ascending limb of loop of Henle. In this review, we provide an overview of the multifaceted physiology and pathophysiology of uromodulin including recent advances in its genetics; cellular trafficking; and mechanistic and clinical studies undertaken to understand the complex relationship between uromodulin, blood pressure, and kidney function. We focus on tubular sodium reabsorption as one of the best understood and pathophysiologically and clinically most important roles of uromodulin, which can lead to therapeutic interventions.
Collapse
Affiliation(s)
- Sheon Mary
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Philipp Boder
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Sandosh Padmanabhan
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Martin W. McBride
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Delyth Graham
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Christian Delles
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| | - Anna F. Dominiczak
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
70
|
Abstract
Recent compelling results indicate possible links between neurotransmitters, intestinal mucosal IgA
+
B cell responses, and immunoglobulin A nephropathy (IgAN) pathogenesis. Here, we demonstrated that γ-amino butyric acid (GABA) transporter-2 (GAT-2) deficiency induces intestinal germinal center (GC) B cell differentiation and worsens the symptoms of IgAN in a mouse model. Mechanistically, GAT-2 deficiency enhances GC B cell differentiation through activation of GABA–mammalian target of rapamycin complex 1 (mTORC1) signaling. In addition, IgAN patients have lower GAT-2 expression but higher activation of mTORC1 in blood B cells, and both are correlated with kidney function in IgAN patients. Collectively, this study describes GABA signaling–mediated intestinal mucosal immunity as a previously unstudied pathogenesis mechanism of IgAN and challenges the current paradigms of IgAN.
Collapse
|
71
|
Khattab A, Torkamani A. Nidogen-1 could play a role in diabetic kidney disease development in type 2 diabetes: a genome-wide association meta-analysis. Hum Genomics 2022; 16:47. [PMID: 36271454 PMCID: PMC9587571 DOI: 10.1186/s40246-022-00422-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Diabetic kidney disease (DKD) affects about 40% of patients with diabetes. It is incurable and usually leads to end-stage renal disease (ESRD). The pathogenesis of DKD is still not fully understood, and the genetics of DKD have not yet been extensively studied. In this study, we investigate the genetic basis of DKD in type 2 diabetes (T2D) to provide more insights into the pathogenesis of the disease. RESULTS Using the data provided by the UK Biobank (UKBB), we performed a DKD genome-wide association study (GWAS) in 13,123 individuals with T2D as well as two creatinine estimated glomerular filtration rate (eGFR) GWA studies: one in 26,786 individuals with T2D and the other in 339,080 non-diabetic individuals. We also conducted a DKD GWAS meta-analysis combining our results with those published by the surrogate markers for micro- and macro-vascular hard endpoints for Innovative diabetes Tools (SUMMIT) consortium. We confirm two loci previously reported to be associated with chronic kidney disease (CKD) and eGFR in T2D. The UMOD-PDILT locus is associated with DKD (P = 1.17E-09) as well as creatinine eGFR in both people with T2D (P = 1.31E-15) and people without diabetes (P = 3.95E-73). The PRKAG2 locus is associated with creatinine eGFR in people with (P = 2.78E-10) and without (P = 5.65E-72) T2D. Our meta-analysis reveals a novel association between DKD and variant rs72763500 (chr1:236116561) which is a splicing quantitative trait locus (sQTL) for nidogen-1 (NID1) gene. CONCLUSION Our data confirm two loci previously reported in association with CKD and creatinine eGFR in T2D. It also suggests that NID1, a major component of the renal tubular basement membrane, could play a role in DKD development in T2D. While our NID1 finding remains to be replicated, it is a step toward a more comprehensive understanding of DKD pathogenesis.
Collapse
Affiliation(s)
- Ahmed Khattab
- Integrative Structural and Computational Biology, Scripps Research, 3344 North Torrey Pines Court, Suite 300, La Jolla, CA, 92037, USA.,Scripps Research Translational Institute, La Jolla, CA, 92037, USA
| | - Ali Torkamani
- Integrative Structural and Computational Biology, Scripps Research, 3344 North Torrey Pines Court, Suite 300, La Jolla, CA, 92037, USA. .,Scripps Research Translational Institute, La Jolla, CA, 92037, USA.
| |
Collapse
|
72
|
Coding Variants of the FMO3 Gene Are Associated with the Risk of Chronic Kidney Disease: A Case-Control Study. Rep Biochem Mol Biol 2022; 11:430-439. [PMID: 36718298 PMCID: PMC9883036 DOI: 10.52547/rbmb.11.3.430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/08/2022] [Indexed: 01/19/2023]
Abstract
Background Chronic kidney disease (CKD) is a global health concern involving roughly one-tenth of developed countries' populations. The flavin-containing dimethylaniline monooxygenase 3 (FMO3) gene encodes an enzyme that catalyzes trimethylamine N-oxide (TMAO), a toxin in CKD sufferers. This preliminary study aims to evaluate the association between coding region variations of FMO3, rs2266782G/A (E158K), rs2266780A/G (E308G), and rs1736557G/A (V257M), and the susceptibility to CKD. Methods A total of 356 participants were enrolled, including 157 patients diagnosed with CKD and 199 age-matched healthy individuals. Genotyping of FMO3 gene variations was performed via PCR-RFLP and ARMS-PCR methods. Results Our findings revealed a significant association between rs2266780A/G and rs1736557G/A and CKD under different genetic models. Compared to the GGG haplotype of rs2266782/rs1736557/rs2266780, the GAG, GAA, AAG, and AAA haplotype combinations conferred an increased risk of CKD in our population. Interaction analysis revealed that some genotype combinations, including GA/AA/AA, AA/AA/AA, GA/AA/GA, and GG/AG/AA, dramatically increased CKD risk in the Iranian population. No correlation was found between FMO3 polymorphisms and CKD stages. Discussion These observations highlight the potential impact of coding variants of the FMO3 gene on the onset of CKD. Further investigations into expanded populations and diverse races are needed to confirm our findings.
Collapse
|
73
|
Simeone CA, Wilkerson JL, Poss AM, Banks JA, Varre JV, Guevara JL, Hernandez EJ, Gorsi B, Atkinson DL, Turapov T, Frodsham SG, Morales JCF, O'Neil K, Moore B, Yandell M, Summers SA, Krolewski AS, Holland WL, Pezzolesi MG. A dominant negative ADIPOQ mutation in a diabetic family with renal disease, hypoadiponectinemia, and hyperceramidemia. NPJ Genom Med 2022; 7:43. [PMID: 35869090 PMCID: PMC9307825 DOI: 10.1038/s41525-022-00314-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 07/06/2022] [Indexed: 01/26/2023] Open
Abstract
Adiponectin, encoded by ADIPOQ, is an insulin-sensitizing, anti-inflammatory, and renoprotective adipokine that activates receptors with intrinsic ceramidase activity. We identified a family harboring a 10-nucleotide deletion mutation in ADIPOQ that cosegregates with diabetes and end-stage renal disease. This mutation introduces a frameshift in exon 3, resulting in a premature termination codon that disrupts translation of adiponectin's globular domain. Subjects with the mutation had dramatically reduced circulating adiponectin and increased long-chain ceramides levels. Functional studies suggest that the mutated protein acts as a dominant negative through its interaction with non-mutated adiponectin, decreasing circulating adiponectin levels, and correlating with metabolic disease.
Collapse
Affiliation(s)
- Christopher A Simeone
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Joseph L Wilkerson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Annelise M Poss
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - James A Banks
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Joseph V Varre
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Jose Lazaro Guevara
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Edgar Javier Hernandez
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Bushra Gorsi
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Donald L Atkinson
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Tursun Turapov
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Scott G Frodsham
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Julio C Fierro Morales
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA
| | - Kristina O'Neil
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, 02115, USA
| | - Barry Moore
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Mark Yandell
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
- Utah Center for Genetic Discovery, Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA
| | - Scott A Summers
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Andrzej S Krolewski
- Section on Genetics and Epidemiology, Research Division, Joslin Diabetes Center, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - William L Holland
- Department of Nutrition and Integrative Physiology, University of Utah College of Health, Salt Lake City, UT, 84112, USA
| | - Marcus G Pezzolesi
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, 84112, USA.
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, 84132, USA.
- Diabetes and Metabolism Research Center, University of Utah School of Medicine, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
74
|
Xu NY, Liu ZY, Yang QM, Bian PP, Li M, Zhao X. Genomic Analyses for Selective Signatures and Genes Involved in Hot Adaptation Among Indigenous Chickens From Different Tropical Climate Regions. Front Genet 2022; 13:906447. [PMID: 35979430 PMCID: PMC9377314 DOI: 10.3389/fgene.2022.906447] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
Climate change, especially weather extremes like extreme cold or extreme hot, is a major challenge for global livestock. One of the animal breeding goals for sustainable livestock production should be to breed animals with excellent climate adaptability. Indigenous livestock and poultry are well adapted to the local climate, and they are good resources to study the genetic footprints and mechanism of the resilience to weather extremes. In order to identify selection signatures and genes that might be involved in hot adaptation in indigenous chickens from different tropical climates, we conducted a genomic analysis of 65 indigenous chickens that inhabit different climates. Several important unique positively selected genes (PSGs) were identified for each local chicken group by the cross-population extended haplotype homozygosity (XP-EHH). These PSGs, verified by composite likelihood ratio, genetic differentiation index, nucleotide diversity, Tajima’s D, and decorrelated composite of multiple signals, are related to nerve regulation, vascular function, immune function, lipid metabolism, kidney development, and function, which are involved in thermoregulation and hot adaptation. However, one common PSG was detected for all three tropical groups of chickens via XP-EHH but was not confirmed by other five types of selective sweep analyses. These results suggest that the hot adaptability of indigenous chickens from different tropical climate regions has evolved in parallel by taking different pathways with different sets of genes. The results from our study have provided reasonable explanations and insights for the rapid adaptation of chickens to diverse tropical climates and provide practical values for poultry breeding.
Collapse
Affiliation(s)
- Nai-Yi Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Zhen-Yu Liu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Qi-Meng Yang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Pei-Pei Bian
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Ming Li
- Department of Biology, University of Konstanz, Konstanz, Germany
| | - Xin Zhao
- Department of Animal Science, McGill University, Montreal, QC, Canada
- *Correspondence: Xin Zhao,
| |
Collapse
|
75
|
Rinschen MM, Palygin O, El-Meanawy A, Domingo-Almenara X, Palermo A, Dissanayake LV, Golosova D, Schafroth MA, Guijas C, Demir F, Jaegers J, Gliozzi ML, Xue J, Hoehne M, Benzing T, Kok BP, Saez E, Bleich M, Himmerkus N, Weisz OA, Cravatt BF, Krüger M, Benton HP, Siuzdak G, Staruschenko A. Accelerated lysine metabolism conveys kidney protection in salt-sensitive hypertension. Nat Commun 2022; 13:4099. [PMID: 35835746 PMCID: PMC9283537 DOI: 10.1038/s41467-022-31670-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 06/27/2022] [Indexed: 01/07/2023] Open
Abstract
Hypertension and kidney disease have been repeatedly associated with genomic variants and alterations of lysine metabolism. Here, we combined stable isotope labeling with untargeted metabolomics to investigate lysine's metabolic fate in vivo. Dietary 13C6 labeled lysine was tracked to lysine metabolites across various organs. Globally, lysine reacts rapidly with molecules of the central carbon metabolism, but incorporates slowly into proteins and acylcarnitines. Lysine metabolism is accelerated in a rat model of hypertension and kidney damage, chiefly through N-alpha-mediated degradation. Lysine administration diminished development of hypertension and kidney injury. Protective mechanisms include diuresis, further acceleration of lysine conjugate formation, and inhibition of tubular albumin uptake. Lysine also conjugates with malonyl-CoA to form a novel metabolite Nε-malonyl-lysine to deplete malonyl-CoA from fatty acid synthesis. Through conjugate formation and excretion as fructoselysine, saccharopine, and Nε-acetyllysine, lysine lead to depletion of central carbon metabolites from the organism and kidney. Consistently, lysine administration to patients at risk for hypertension and kidney disease inhibited tubular albumin uptake, increased lysine conjugate formation, and reduced tricarboxylic acid (TCA) cycle metabolites, compared to kidney-healthy volunteers. In conclusion, lysine isotope tracing mapped an accelerated metabolism in hypertension, and lysine administration could protect kidneys in hypertensive kidney disease.
Collapse
Affiliation(s)
- Markus M Rinschen
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.
- III. Medical Clinic, University Hospital Hamburg Eppendorf, Hamburg, Germany.
- AIAS, Aarhus Institute of Advanced Studies (AIAS), Aarhus University, Aarhus, Denmark.
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Ashraf El-Meanawy
- Division of Nephrology, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Xavier Domingo-Almenara
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Omics Sciences Unit, EURECAT, Technology Centre of Catalonia, Reus, Catalonia, Spain
| | - Amelia Palermo
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, University of California, Los Angeles, USA
| | - Lashodya V Dissanayake
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Daria Golosova
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | | | - Carlos Guijas
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Fatih Demir
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | | | - Megan L Gliozzi
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Jingchuan Xue
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Martin Hoehne
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Thomas Benzing
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
- Department II of Internal Medicine, University Hospital of Cologne, Cologne, Germany
| | - Bernard P Kok
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Enrique Saez
- Department of Molecular Medicine, Scripps Research, La Jolla, CA, 92037, USA
| | - Markus Bleich
- Institute of Physiology, University Kiel, Kiel, Germany
| | | | - Ora A Weisz
- Renal Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | | | - Marcus Krüger
- Center for Molecular Medicine Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, Cologne, Germany
| | - H Paul Benton
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA
| | - Gary Siuzdak
- Scripps Center for Metabolomics, Scripps Research, La Jolla, CA, 92037, USA.
| | - Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, FL, 33602, USA.
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
- James A. Haley Veterans' Hospital, Tampa, FL, 33612, USA.
- Hypertension and Kidney Research Center, University of South Florida, Tampa, FL, 33602, USA.
| |
Collapse
|
76
|
Wang L, Zhou Z, Yang Y, Gao P, Lin X, Wu Z. A Genetic Polymorphism in the WDR72 Gene is Associated With Calcium Nephrolithiasis in the Chinese Han Population. Front Genet 2022; 13:897051. [PMID: 35910217 PMCID: PMC9333346 DOI: 10.3389/fgene.2022.897051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 06/24/2022] [Indexed: 12/02/2022] Open
Abstract
A previous genome-wide association study (GWAS) reported several novel loci for nephrolithiasis in British and Japanese population, some of which were predicted to influence CaSR signaling. In this study, we aimed to evaluate the association of these loci with calcium nephrolithiasis in Chinese Han population. We performed a case-control association analysis involving 691 patients with calcium nephrolithiasis and 1008 control subjects. We were able to genotype a total of 17 single-nucleotide polymorphisms (SNPs), which were previously reported to be significantly associated with nephrolithiasis in GWAS. rs578595 at WDR72 was significantly associated with calcium nephrolithiasis in Chinese Han population (p < 0.001, OR = 0.617). Moreover, rs12654812 at SLC34A1 (p = 0.0427, OR = 1.170), rs12539707 at HIBADH (p = 0.0179, OR = 0.734), rs1037271 at DGKH (p = 0.0096, OR = 0.828) and rs12626330 at CLDN14 (p = 0.0080, OR = 1.213) indicated suggestive associations with calcium nephrolithiasis. Our results elucidated the significance of genetic variation at WDR72, DGKH, CLDN14, SLC34A1, and HIBADH in Chinese patients with nephrolithiasis. Since polymorphisms of WDR72, DGKH, and CLDN14 are predicted to influence in CaSR signaling, our results emphasized the role of abnormal calcium homeostasis in calcium nephrolithiasis.
Collapse
Affiliation(s)
- Lujia Wang
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, China
- Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zijian Zhou
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, China
- Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yuanyuan Yang
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, China
- Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, China
| | - Peng Gao
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, China
- Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoling Lin
- Department of Urology, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xiaoling Lin, ; Zhong Wu,
| | - Zhong Wu
- Department of Urology, Huashan Hospital & Institute of Urology, Fudan University, Shanghai, China
- Clinical Research Center of Urolithiasis, Shanghai Medical College, Fudan University, Shanghai, China
- *Correspondence: Xiaoling Lin, ; Zhong Wu,
| |
Collapse
|
77
|
Liu C, Wang Z, Hui Q, Chiang Y, Chen J, Brijkumar J, Edwards JA, Ordonez CE, Dudgeon MR, Sunpath H, Pillay S, Moodley P, Kuritzkes DR, Moosa MYS, Jones DP, Marconi VC, Sun YV. Crosstalk between Host Genome and Metabolome among People with HIV in South Africa. Metabolites 2022; 12:metabo12070624. [PMID: 35888748 PMCID: PMC9316179 DOI: 10.3390/metabo12070624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/16/2022] [Accepted: 07/01/2022] [Indexed: 02/01/2023] Open
Abstract
Genome-wide association studies (GWAS) of circulating metabolites have revealed the role of genetic regulation on the human metabolome. Most previous investigations focused on European ancestry, and few studies have been conducted among populations of African descent living in Africa, where the infectious disease burden is high (e.g., human immunodeficiency virus (HIV)). It is important to understand the genetic associations of the metabolome in diverse at-risk populations including people with HIV (PWH) living in Africa. After a thorough literature review, the reported significant gene−metabolite associations were tested among 490 PWH in South Africa. Linear regression was used to test associations between the candidate metabolites and genetic variants. GWAS of 154 plasma metabolites were performed to identify novel genetic associations. Among the 29 gene−metabolite associations identified in the literature, we replicated 10 in South Africans with HIV. The UGT1A cluster was associated with plasma levels of biliverdin and bilirubin; SLC16A9 and CPS1 were associated with carnitine and creatine, respectively. We also identified 22 genetic associations with metabolites using a genome-wide significance threshold (p-value < 5 × 10−8). In a GWAS of plasma metabolites in South African PWH, we replicated reported genetic associations across ancestries, and identified novel genetic associations using a metabolomics approach.
Collapse
Affiliation(s)
- Chang Liu
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Zicheng Wang
- College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA;
| | - Qin Hui
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Yiyun Chiang
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Junyu Chen
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
| | - Jaysingh Brijkumar
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Johnathan A. Edwards
- Department of Biostatistics and Bioinformatics, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
- Lincoln International Institute for Rural Health, School of Health and Social Care, University of Lincoln, Lincoln LN6 7TS, UK
| | - Claudia E. Ordonez
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
| | - Mathew R. Dudgeon
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
| | - Henry Sunpath
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Selvan Pillay
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Pravi Moodley
- National Health Laboratory Service, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4011, South Africa;
| | - Daniel R. Kuritzkes
- Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA;
| | - Mohamed Y. S. Moosa
- Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban 4041, South Africa; (J.B.); (H.S.); (S.P.); (M.Y.S.M.)
| | - Dean P. Jones
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
| | - Vincent C. Marconi
- School of Medicine, Emory University, Atlanta, GA 30322, USA; (M.R.D.); (D.P.J.); (V.C.M.)
- Hubert Department of Global Health, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA;
- Emory Vaccine Center, Atlanta, GA 30322, USA
| | - Yan V. Sun
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA 30322, USA; (C.L.); (Q.H.); (Y.C.); (J.C.)
- Correspondence: ; Tel.: +1-404-727-9090
| |
Collapse
|
78
|
Kopp JB, Heymann J. Impact of APOL1 kidney risk variants on glomerular transcriptomes. Kidney Int 2022; 102:16-19. [PMID: 35738828 DOI: 10.1016/j.kint.2022.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 04/20/2022] [Accepted: 04/27/2022] [Indexed: 11/29/2022]
Abstract
McNulty and colleagues describe the glomerular transcriptional landscape of subjects with APOL1 (the gene encoding apolipoprotein L1)-associated kidney disease, using bulk RNA sequencing. They found the following: APOL1 gene expression was higher in individuals with APOL1 high-risk genetic status; in glomeruli, STC1, encoding stanniocalcin, was the most upregulated gene, and CCL18, encoding C-C motif chemokine ligand 18, was the most downregulated gene; and nuclear factor kappa BNF-κB inhibitor-interacting Ras-like 1 (NKIRAS1) is the strongest hub gene. These findings identify disease pathways that might mediate or mitigate APOL1-associated nephropathies.
Collapse
|
79
|
Jiang M, Wang X, Gao X, Cardenas A, Baccarelli AA, Guo X, Huang J, Wu S. Association of DNA methylation in circulating CD4 +T cells with short-term PM 2.5 pollution waves: A quasi-experimental study of healthy young adults. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113634. [PMID: 35617899 DOI: 10.1016/j.ecoenv.2022.113634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/15/2022] [Accepted: 05/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Fine particulate matter (PM2.5) is a modifiable environmental risk factor with established adverse effects on human health. However, associations between acute PM2.5 fluctuation and DNA methylation remain unknown. METHODS A quasi-experimental study utilizing naturally occurring PM2.5 pollution waves (PPWs) was conducted on 32 healthy young adults. Repeated follow-up measurements were performed and participants served as their own controls before, during, and after PPWs. Exposure measurements including indoor and ambient PM2.5 levels, and equivalent personal PM2.5 exposure were further estimated based on the time-location information. DNA methylation profiles of circulating CD4+T cells were obtained using Illumina HumanMethylationEPIC BeadChip. Linear mixed-effect models were applied to estimate the associations between two scenarios (during-PPWs vs. pre-PPWs periods and during-PPWs vs. post-PPWs periods) and methylation level of each CpG site. We further validated their associations with the personal PM2.5 exposure, and GO and KEGG analyses and mediation analysis were conducted accordingly. RESULTS Data from 26 participants were included in final analysis after quality control. Short-term high PM2.5 exposure was associated with DNA methylation changes of participants. Nine differently methylated CpG sites were not only significantly associated with PPWs periods but also with personal PM2.5 exposure in 24-h prior to the health examinations (p < 0.01). Gene ontology analysis found that five sites were associated with two pathways relating to membrane protein synthesis. PM2.5-related changes in CpG sites were mediated by sP-selectin, 8-isoPGF2α, EGF, GRO, IL-15, and IFN-α2, with mediated proportions ranging from 9.65% to 23.40%. CONCLUSIONS This is the first quasi-experimental study showing that short-term high PM2.5 exposure could alter the DNA methylation of CD4+T cells, which provided valuable information for further exploring underlying biological mechanisms and epigenetic biomarkers for PM2.5-related acute health effects.
Collapse
Affiliation(s)
- Meijie Jiang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Xinmei Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Xu Gao
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Andres Cardenas
- Division of Environmental Health Sciences, School of Public Health and Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Xinbiao Guo
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Jing Huang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China.
| | - Shaowei Wu
- Department of Occupational and Environmental Health, School of Public Health, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, China; Key Laboratory of Trace Elements and Endemic Diseases in Ministry of Health, Xi'an, Shaanxi, China; Key Laboratory for Disease Prevention and Control and Health Promotion of Shaanxi Province, Xi'an, Shaanxi, China.
| |
Collapse
|
80
|
Rysz J, Franczyk B, Rysz-Górzyńska M, Gluba-Brzózka A. Are Alterations in DNA Methylation Related to CKD Development? Int J Mol Sci 2022; 23:7108. [PMID: 35806113 PMCID: PMC9267048 DOI: 10.3390/ijms23137108] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/17/2022] [Accepted: 06/18/2022] [Indexed: 12/29/2022] Open
Abstract
The modifications in genomic DNA methylation are involved in the regulation of normal and pathological cellular processes. The epigenetic regulation stimulates biological plasticity as an adaptive response to variations in environmental factors. The role of epigenetic changes is vital for the development of some diseases, including atherogenesis, cancers, and chronic kidney disease (CKD). The results of studies presented in this review have suggested that altered DNA methylation can modulate the expression of pro-inflammatory and pro-fibrotic genes, as well those essential for kidney development and function, thus stimulating renal disease progression. Abnormally increased homocysteine, hypoxia, and inflammation have been suggested to alter epigenetic regulation of gene expression in CKD. Studies of renal samples have demonstrated the relationship between variations in DNA methylation and fibrosis and variations in estimated glomerular filtration rate (eGFR) in human CKD. The unravelling of the genetic-epigenetic profile would enhance our understanding of processes underlying the development of CKD. The understanding of multifaceted relationship between DNA methylation, genes expression, and disease development and progression could improve the ability to identify individuals at risk of CKD and enable the choice of appropriate disease management.
Collapse
Affiliation(s)
- Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskego Street, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskego Street, 90-549 Lodz, Poland; (J.R.); (B.F.)
| | - Magdalena Rysz-Górzyńska
- Department of Otolaryngology, Laryngological Oncology, Audiology and Phoniatrics, Medical Univesity of Lodz, 113 Żeromskego Street, 90-549 Lodz, Poland;
| | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskego Street, 90-549 Lodz, Poland; (J.R.); (B.F.)
| |
Collapse
|
81
|
Genetics in chronic kidney disease: conclusions from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int 2022; 101:1126-1141. [PMID: 35460632 PMCID: PMC9922534 DOI: 10.1016/j.kint.2022.03.019] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/16/2022] [Accepted: 03/29/2022] [Indexed: 01/19/2023]
Abstract
Numerous genes for monogenic kidney diseases with classical patterns of inheritance, as well as genes for complex kidney diseases that manifest in combination with environmental factors, have been discovered. Genetic findings are increasingly used to inform clinical management of nephropathies, and have led to improved diagnostics, disease surveillance, choice of therapy, and family counseling. All of these steps rely on accurate interpretation of genetic data, which can be outpaced by current rates of data collection. In March of 2021, Kidney Diseases: Improving Global Outcomes (KDIGO) held a Controversies Conference on "Genetics in Chronic Kidney Disease (CKD)" to review the current state of understanding of monogenic and complex (polygenic) kidney diseases, processes for applying genetic findings in clinical medicine, and use of genomics for defining and stratifying CKD. Given the important contribution of genetic variants to CKD, practitioners with CKD patients are advised to "think genetic," which specifically involves obtaining a family history, collecting detailed information on age of CKD onset, performing clinical examination for extrarenal symptoms, and considering genetic testing. To improve the use of genetics in nephrology, meeting participants advised developing an advanced training or subspecialty track for nephrologists, crafting guidelines for testing and treatment, and educating patients, students, and practitioners. Key areas of future research, including clinical interpretation of genome variation, electronic phenotyping, global representation, kidney-specific molecular data, polygenic scores, translational epidemiology, and open data resources, were also identified.
Collapse
|
82
|
Li Y, Cheng Y, Consolato F, Schiano G, Chong MR, Pietzner M, Nguyen NQH, Scherer N, Biggs ML, Kleber ME, Haug S, Göçmen B, Pigeyre M, Sekula P, Steinbrenner I, Schlosser P, Joseph CB, Brody JA, Grams ME, Hayward C, Schultheiss UT, Krämer BK, Kronenberg F, Peters A, Seissler J, Steubl D, Then C, Wuttke M, März W, Eckardt KU, Gieger C, Boerwinkle E, Psaty BM, Coresh J, Oefner PJ, Pare G, Langenberg C, Scherberich JE, Yu B, Akilesh S, Devuyst O, Rampoldi L, Köttgen A. Genome-wide studies reveal factors associated with circulating uromodulin and its relationships to complex diseases. JCI Insight 2022; 7:e157035. [PMID: 35446786 PMCID: PMC9220927 DOI: 10.1172/jci.insight.157035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 04/07/2022] [Indexed: 11/28/2022] Open
Abstract
Uromodulin (UMOD) is a major risk gene for monogenic and complex forms of kidney disease. The encoded kidney-specific protein uromodulin is highly abundant in urine and related to chronic kidney disease, hypertension, and pathogen defense. To gain insights into potential systemic roles, we performed genome-wide screens of circulating uromodulin using complementary antibody-based and aptamer-based assays. We detected 3 and 10 distinct significant loci, respectively. Integration of antibody-based results at the UMOD locus with functional genomics data (RNA-Seq, ATAC-Seq, Hi-C) of primary human kidney tissue highlighted an upstream variant with differential accessibility and transcription in uromodulin-synthesizing kidney cells as underlying the observed cis effect. Shared association patterns with complex traits, including chronic kidney disease and blood pressure, placed the PRKAG2 locus in the same pathway as UMOD. Experimental validation of the third antibody-based locus, B4GALNT2, showed that the p.Cys466Arg variant of the encoded N-acetylgalactosaminyltransferase had a loss-of-function effect leading to higher serum uromodulin levels. Aptamer-based results pointed to enzymes writing glycan marks present on uromodulin and to their receptors in the circulation, suggesting that this assay permits investigating uromodulin's complex glycosylation rather than its quantitative levels. Overall, our study provides insights into circulating uromodulin and its emerging functions.
Collapse
Affiliation(s)
- Yong Li
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Francesco Consolato
- Molecular Genetics of Renal Disorders group, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Michael R. Chong
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences and
- Department of Pathology and Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Maik Pietzner
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Ngoc Quynh H. Nguyen
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Nora Scherer
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Spemann Graduate School of Biology and Medicine, University of Freiburg, Freiburg, Germany
| | - Mary L. Biggs
- Cardiovascular Health Research Unit, Department of Medicine, and
- Department of Biostatistics, University of Washington, Seattle, Washington, USA
| | - Marcus E. Kleber
- SYNLAB MVZ Humangenetik Mannheim GmbH, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Stefan Haug
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Burulça Göçmen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Marie Pigeyre
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Medicine, Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Peggy Sekula
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Inga Steinbrenner
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
| | - Pascal Schlosser
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Christina B. Joseph
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | | | - Morgan E. Grams
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Division of Nephrology, School of Medicine, Johns Hopkins University, Baltimore, Maryland, USA
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Western General Hospital, Edinburgh, United Kingdom
| | - Ulla T. Schultheiss
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Medicine IV: Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Bernhard K. Krämer
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Annette Peters
- Institute of Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Chair of Epidemiology, Institute for Medical Information Processing, Biometry, and Epidemiology, Faculty of Medicine, Ludwig-Maximilians-Universität (LMU), Munich, Germany
| | - Jochen Seissler
- Medical Clinic and Policlinic IV, Hospital of the University of Munich, LMU Munich, Munich, Germany
| | - Dominik Steubl
- Division of Nephrology, Tufts Medical Center, Boston, Massachusetts, USA
- Department of Nephrology, Klinikum rechts der Isar, Technical University Munich, Munich, Germany
- Boehringer Ingelheim International GmbH, Ingelheim, Germany
| | - Cornelia Then
- Medical Clinic and Policlinic IV, Hospital of the University of Munich, LMU Munich, Munich, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Medicine IV: Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Winfried März
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Graz, Austria
- SYNLAB Academy, SYNLAB Holding Deutschland GmbH, Augsburg and Mannheim, Germany
| | - Kai-Uwe Eckardt
- Department of Nephrology and Medical Intensive Care, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Partner Munich, Neuherberg, Germany
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, and
- Department of Epidemiology and
- Department of Health Systems and Population Health, School of Public Health, University of Washington, Seattle, Washington, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Peter J. Oefner
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Guillaume Pare
- Population Health Research Institute and Thrombosis and Atherosclerosis Research Institute, David Braley Cardiac, Vascular and Stroke Research Institute, Hamilton Health Sciences, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, Faculty of Health Science, McMaster University, Hamilton, Ontario, Canada
| | - Claudia Langenberg
- Medical Research Council (MRC) Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Computational Medicine, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
| | | | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Shreeram Akilesh
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Luca Rampoldi
- Molecular Genetics of Renal Disorders group, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, and
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
- Centre for Integrative Biological Signalling Studies (CIBSS), University of Freiburg, Freiburg, Germany
| |
Collapse
|
83
|
Tariq A, Chen J, Yu B, Boerwinkle E, Coresh J, Grams ME, Rebholz CM. Metabolomics of Dietary Acid Load and Incident Chronic Kidney Disease. J Ren Nutr 2022; 32:292-300. [PMID: 34294549 PMCID: PMC8766597 DOI: 10.1053/j.jrn.2021.05.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 04/29/2021] [Accepted: 05/15/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Blood biomarkers of dietary intake are more objective than self-reported dietary intake. Metabolites associated with dietary acid load were previously identified in 2 chronic kidney disease (CKD) populations. We aimed to extend these findings to a general population, replicating their association with dietary acid load, and investigating whether the individual biomarkers were prospectively associated with incident CKD. METHODS Among 15,792 participants in the Atherosclerosis Risk in Communities cohort followed up from 1987 to 1989 (baseline) to 2019, we evaluated 3,844 black and white men and women with dietary and metabolomic data in cross-sectional and prospective analyses. We hypothesized that a higher dietary acid load (using equations for potential renal acid load and net endogenous acid production) was associated with lower serum levels of 12 previously identified metabolites: indolepropionylglycine, indolepropionate, N-methylproline, N-δ-acetylornithine, threonate, oxalate, chiro-inositol, methyl glucopyranoside, stachydrine, catechol sulfate, hippurate, and tartronate. In addition, we hypothesized that lower serum levels of these 12 metabolites were associated with higher risk of incident CKD. RESULTS Eleven out of 12 metabolites were significantly inversely associated with dietary acid load, after adjusting for demographics, socioeconomic status, health behaviors, health status, and estimated glomerular filtration rate: indolepropionylglycine, indolepropionate, N-methylproline, threonate, oxalate, chiro-inositol, catechol sulfate, hippurate, methyl glucopyranoside (α + β), stachydrine, and tartronate. N-methylproline was inversely associated with incident CKD (hazard ratio: 0.95, 95% confidence interval: 0.91, 0.99, P = .01). The metabolomic biomarkers of dietary acid load significantly improved prediction of elevated dietary acid load estimated using dietary data, beyond covariates (difference in C statistics: 0.021-0.077, P ≤ 1.08 × 10-3). CONCLUSION Inverse associations between candidate biomarkers of dietary acid load were replicated in a general population. N-methylproline, representative of citrus fruit consumption, is a promising marker of dietary acid load and could represent an important pathway between dietary acid load and CKD.
Collapse
Affiliation(s)
- Anam Tariq
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jingsha Chen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas Health Science Center at Houston, Houston, Texas
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Morgan E Grams
- Division of Nephrology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Casey M Rebholz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland.
| |
Collapse
|
84
|
Tayyeb A, Dihazi GH, Tampe B, Zeisberg M, Tampe D, Hakroush S, Bührig C, Frese J, Serin N, Eltoweissy M, Müller GA, Dihazi H. Calreticulin Shortage Results in Disturbance of Calcium Storage, Mitochondrial Disease, and Kidney Injury. Cells 2022; 11:cells11081329. [PMID: 35456008 PMCID: PMC9025518 DOI: 10.3390/cells11081329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 12/10/2022] Open
Abstract
Renal Ca2+ reabsorption plays a central role in the fine-tuning of whole-body Ca2+ homeostasis. Here, we identified calreticulin (Calr) as a missing link in Ca2+ handling in the kidney and showed that a shortage of Calr results in mitochondrial disease and kidney pathogenesis. We demonstrated that Calr+/− mice displayed a chronic physiological low level of Calr and that this was associated with progressive renal injury manifested in glomerulosclerosis and tubulointerstitial damage. We found that Calr+/− kidney cells suffer from a disturbance in functionally active calcium stores and decrease in Ca2+ storage capacity. Consequently, the kidney cells displayed an abnormal activation of Ca2+ signaling and NF-κB pathways, resulting in inflammation and wide progressive kidney injury. Interestingly, the disturbance in the Ca2+ homeostasis and signaling in Calr+/− kidney mice cells triggered severe mitochondrial disease and aberrant mitophagy, resulting in a high level of oxidative stress and energy shortage. These findings provide novel mechanistic insight into the role of Calr in kidney calcium handling, function, and pathogenesis.
Collapse
Affiliation(s)
- Asima Tayyeb
- School of Biological Sciences, University of the Punjab, Lahore 53700, Pakistan;
| | - Gry H. Dihazi
- UMG-Laboratories, Institute for Clinical Chemistry, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany;
| | - Björn Tampe
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
| | - Michael Zeisberg
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
| | - Desiree Tampe
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
| | - Samy Hakroush
- Department of Pathology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany;
| | - Charlotte Bührig
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
| | - Jenny Frese
- Department of Occupational Medicine and Health Safety, Deutsche Post AG, Kölnische Strasse 81, 34117 Kassel, Germany;
| | - Nazli Serin
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
- Department of Hematology and Oncology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany
| | - Marwa Eltoweissy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria 21568, Egypt;
| | - Gerhard A. Müller
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
| | - Hassan Dihazi
- Clinic for Nephrology and Rheumatology, University Medical Centre Göttingen, Robert-Koch-Strasse 40, 37075 Göttingen, Germany; (B.T.); (M.Z.); (D.T.); (C.B.); (N.S.); (G.A.M.)
- Centre for Biostructural Imaging of Neurodegeneration (BIN), University Medical Center Göttingen, 37075 Göttingen, Germany
- Correspondence: ; Tel.: +49-551-3960350
| |
Collapse
|
85
|
Rhee EP, Surapaneni A, Zheng Z, Zhou L, Dutta D, Arking DE, Zhang J, Duong T, Chatterjee N, Luo S, Schlosser P, Mehta R, Waikar SS, Saraf SL, Kelly TN, Hamm LL, Rao PS, Mathew AV, Hsu CY, Parsa A, Vasan RS, Kimmel PL, Clish CB, Coresh J, Feldman HI, Grams ME. Trans-ethnic genome-wide association study of blood metabolites in the Chronic Renal Insufficiency Cohort (CRIC) study. Kidney Int 2022; 101:814-823. [PMID: 35120996 PMCID: PMC8940669 DOI: 10.1016/j.kint.2022.01.014] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 12/06/2021] [Accepted: 01/11/2022] [Indexed: 12/14/2022]
Abstract
Metabolomics genome wide association study (GWAS) help outline the genetic contribution to human metabolism. However, studies to date have focused on relatively healthy, population-based samples of White individuals. Here, we conducted a GWAS of 537 blood metabolites measured in the Chronic Renal Insufficiency Cohort (CRIC) Study, with separate analyses in 822 White and 687 Black study participants. Trans-ethnic meta-analysis was then applied to improve fine-mapping of potential causal variants. Mean estimated glomerular filtration rate was 44.4 and 41.5 mL/min/1.73m2 in the White and Black participants, respectively. There were 45 significant metabolite associations at 19 loci, including novel associations at PYROXD2, PHYHD1, FADS1-3, ACOT2, MYRF, FAAH, and LIPC. The strength of associations was unchanged in models additionally adjusted for estimated glomerular filtration rate and proteinuria, consistent with a direct biochemical effect of gene products on associated metabolites. At several loci, trans-ethnic meta-analysis, which leverages differences in linkage disequilibrium across populations, reduced the number and/or genomic interval spanned by potentially causal single nucleotide polymorphisms compared to fine-mapping in the White participant cohort alone. Across all validated associations, we found strong concordance in effect sizes of the potentially causal single nucleotide polymorphisms between White and Black study participants. Thus, our study identifies novel genetic determinants of blood metabolites in chronic kidney disease, demonstrates the value of diverse cohorts to improve causal inference in metabolomics GWAS, and underscores the shared genetic basis of metabolism across race.
Collapse
Affiliation(s)
- Eugene P Rhee
- Nephrology Division and Endocrine Unit, Massachusetts General Hospital, Boston, Massachussetts, USA.
| | - Aditya Surapaneni
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Zihe Zheng
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Linda Zhou
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Diptavo Dutta
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Dan E Arking
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jingning Zhang
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - ThuyVy Duong
- McKusick-Nathans Institute, Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nilanjan Chatterjee
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Shengyuan Luo
- Department of Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Pascal Schlosser
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Rupal Mehta
- Division of Nephrology and Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Sushrut S Waikar
- Section of Nephrology, Boston University School of Medicine, Boston Medical Center, Boston, Massachussetts, USA
| | - Santosh L Saraf
- Division of Hematology and Oncology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Tanika N Kelly
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Lee L Hamm
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Panduranga S Rao
- Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Anna V Mathew
- Division of Nephrology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Chi-Yuan Hsu
- Division of Nephrology, University of California, San Francisco School of Medicine, San Francisco, California, USA
| | - Afshin Parsa
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ramachandran S Vasan
- Section of Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, Massachussetts, USA; Section of Cardiology, Department of Medicine, Boston University School of Medicine, Boston, Massachussetts, USA; Department of Epidemiology, Boston University School of Public Health, Boston, Massachussetts, USA
| | - Paul L Kimmel
- Division of Kidney, Urologic, and Hematologic Diseases, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Clary B Clish
- Broad Institute of MIT and Harvard, Cambridge, Massachussetts, USA
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA
| | - Harold I Feldman
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Morgan E Grams
- Department of Epidemiology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA; Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University, Baltimore, Maryland, USA; Division of Nephrology, Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.
| |
Collapse
|
86
|
Yeh KH, Hsu LA, Teng MS, Wu S, Chou HH, Ko YL. Pleiotropic Effects of Common and Rare GCKR Exonic Mutations on Cardiometabolic Traits. Genes (Basel) 2022; 13:genes13030491. [PMID: 35328045 PMCID: PMC8951277 DOI: 10.3390/genes13030491] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/04/2022] [Accepted: 03/06/2022] [Indexed: 02/05/2023] Open
Abstract
Background: The common non-synonymous mutation of the glucokinase regulator (GCKR) gene, namely rs1260326, is widely reported to have pleiotropic effects on cardio-metabolic traits and hematological parameters. Objective: This study aimed to identify whether other GCKR variants may have pleiotropic effects independent of the rs1260326 genotypes. Methods: In total, 81,097 Taiwan Biobank participants were enrolled for the regional plot association studies and candidate variant analysis of the region around the GCKR gene. Results: The initial candidate variant approach showed the significant association of the rs1260326 genotypes with multiple phenotypes. Regional plot association analysis of the GCKR gene region further revealed genome-wide significant associations between GCKR variants and serum total and low-density lipoprotein cholesterol; triglyceride, uric acid, creatinine, aspartate aminotransferase, γ-Glutamyl transferase, albumin, and fasting plasma glucose levels; estimated glomerular filtration rate; leukocyte and platelet counts; microalbuminuria, and metabolic syndrome, with rs1260326 being the most common lead polymorphism. Serial conditional analysis identified genome-wide significant associations of two low-frequency exonic mutations, rs143881585 and rs8179206, with high serum triglyceride and albumin levels. In five rare GCKR exonic non-synonymous or nonsense mutations available for analysis, GCKR rs146175795 showed an independent association with serum triglyceride and albumin levels and rs150673460 showed an independent association with serum triglyceride levels. Weighted genetic risk scores from the combination of GCKR rs143881585 and rs146175795 revealed a significant association with metabolic syndrome. Conclusion: In addition to the rs1260326 variant, low-frequency and rare GCKR exonic mutations exhibit pleiotropic effects on serum triglyceride and albumin levels and the risk of metabolic syndrome. These results provide evidence that both common and rare GCKR variants may play a critical role in predicting the risk of cardiometabolic disorders.
Collapse
Affiliation(s)
- Kuan-Hung Yeh
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; (K.-H.Y.); (H.-H.C.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Lung-An Hsu
- The First Cardiovascular Division, Department of Internal Medicine, Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Taoyuan 33305, Taiwan;
| | - Ming-Sheng Teng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan;
| | - Semon Wu
- Department of Life Science, Chinese Culture University, Taipei 11114, Taiwan;
| | - Hsin-Hua Chou
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; (K.-H.Y.); (H.-H.C.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Yu-Lin Ko
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; (K.-H.Y.); (H.-H.C.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan;
- Correspondence: ; Tel.: +886-2-6628-9779 (ext. 5355); Fax: +886-2-6628-9009
| |
Collapse
|
87
|
Zhang H, Mo X, Wang A, Peng H, Guo D, Zhong C, Zhu Z, Xu T, Zhang Y. Association of DNA Methylation in Blood Pressure-Related Genes With Ischemic Stroke Risk and Prognosis. Front Cardiovasc Med 2022; 9:796245. [PMID: 35345488 PMCID: PMC8957103 DOI: 10.3389/fcvm.2022.796245] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 01/31/2022] [Indexed: 12/16/2022] Open
Abstract
BackgroundA genome-wide association study identified 12 genetic loci influencing blood pressure and implicated a role of DNA methylation. However, the relationship between methylation and ischemic stroke has not yet been clarified. We conducted a large-sample sequencing study to identify blood leukocyte DNA methylations as novel biomarkers for ischemic stroke risk and prognosis based on previously identified genetic loci.MethodsMethylation levels of 17 genes were measured by sequencing in 271 ischemic stroke cases and 323 controls, and the significant associations were validated in another independent sample of 852 cases and 925 controls. The associations between methylation levels and ischemic stroke risk and prognosis were evaluated.ResultsMethylation of AMH, C17orf82, HDAC9, IGFBP3, LRRC10B, PDE3A, PRDM6, SYT7 and TBX2 was significantly associated with ischemic stroke. Compared to participants without any hypomethylated targets, the odds ratio (OR) (95% confidence interval, CI) for those with 9 hypomethylated genes was 1.41 (1.33–1.51) for ischemic stroke. Adding methylation levels of the 9 genes to the basic model of traditional risk factors significantly improved the risk stratification for ischemic stroke. Associations between AMH, HDAC9, IGFBP3, PDE3A and PRDM6 gene methylation and modified Rankin Scale scores were significant after adjustment for covariates. Lower methylation levels of AMH, C17orf82, PRDM6 and TBX2 were significantly associated with increased 3-month mortality. Compared to patients without any hypomethylated targets, the OR (95% CI) for those with 4 hypomethylated targets was 1.12 (1.08–1.15) for 3-month mortality (P = 2.28 × 10−10).ConclusionThe present study identified blood leukocyte DNA methylations as potential factors affecting ischemic stroke risk and prognosis among Han Chinese individuals.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Hao Peng
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Daoxia Guo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Chongke Zhong
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Zhengbao Zhu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yonghong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China
- Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
- *Correspondence: Yonghong Zhang
| |
Collapse
|
88
|
Joseph CB, Mariniello M, Yoshifuji A, Schiano G, Lake J, Marten J, Richmond A, Huffman JE, Campbell A, Harris SE, Troyanov S, Cocca M, Robino A, Thériault S, Eckardt KU, Wuttke M, Cheng Y, Corre T, Kolcic I, Black C, Bruat V, Concas MP, Sala C, Aeschbacher S, Schaefer F, Bergmann S, Campbell H, Olden M, Polasek O, Porteous DJ, Deary IJ, Madore F, Awadalla P, Girotto G, Ulivi S, Conen D, Wuehl E, Olinger E, Wilson JF, Bochud M, Köttgen A, Hayward C, Devuyst O. Meta-GWAS Reveals Novel Genetic Variants Associated with Urinary Excretion of Uromodulin. J Am Soc Nephrol 2022; 33:511-529. [PMID: 35228297 PMCID: PMC8975067 DOI: 10.1681/asn.2021040491] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Uromodulin, the most abundant protein excreted in normal urine, plays major roles in kidney physiology and disease. The mechanisms regulating the urinary excretion of uromodulin remain essentially unknown. METHODS We conducted a meta-analysis of genome-wide association studies for raw (uUMOD) and indexed to creatinine (uUCR) urinary levels of uromodulin in 29,315 individuals of European ancestry from 13 cohorts. We tested the distribution of candidate genes in kidney segments and investigated the effects of keratin-40 (KRT40) on uromodulin processing. RESULTS Two genome-wide significant signals were identified for uUMOD: a novel locus (P 1.24E-08) over the KRT40 gene coding for KRT40, a type 1 keratin expressed in the kidney, and the UMOD-PDILT locus (P 2.17E-88), with two independent sets of single nucleotide polymorphisms spread over UMOD and PDILT. Two genome-wide significant signals for uUCR were identified at the UMOD-PDILT locus and at the novel WDR72 locus previously associated with kidney function. The effect sizes for rs8067385, the index single nucleotide polymorphism in the KRT40 locus, were similar for both uUMOD and uUCR. KRT40 colocalized with uromodulin and modulating its expression in thick ascending limb (TAL) cells affected uromodulin processing and excretion. CONCLUSIONS Common variants in KRT40, WDR72, UMOD, and PDILT associate with the levels of uromodulin in urine. The expression of KRT40 affects uromodulin processing in TAL cells. These results, although limited by lack of replication, provide insights into the biology of uromodulin, the role of keratins in the kidney, and the influence of the UMOD-PDILT locus on kidney function.
Collapse
Affiliation(s)
- Christina B Joseph
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Marta Mariniello
- Mechanisms of Inherited Kidney Disorders Group, Institute of Physiology Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Ayumi Yoshifuji
- Mechanisms of Inherited Kidney Disorders Group, Institute of Physiology Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Guglielmo Schiano
- Mechanisms of Inherited Kidney Disorders Group, Institute of Physiology Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Jennifer Lake
- Mechanisms of Inherited Kidney Disorders Group, Institute of Physiology Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Jonathan Marten
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Anne Richmond
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer E Huffman
- Center for Population Genomics,VA Boston Healthcare System, Jamaica Plain, Massachusetts
- The Framingham Heart Study, Framingham, Massachusetts
| | - Archie Campbell
- Centre for Genomic & Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah E Harris
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephan Troyanov
- Division of Nephrology, Hôpital du Sacre-Coeur de Montreal, Montreal, Canada
| | - Massimiliano Cocca
- Institute for Maternal and Child Health IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo" 34127 Trieste, Italy
| | - Antonietta Robino
- Institute for Maternal and Child Health IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo" 34127 Trieste, Italy
| | - Sébastien Thériault
- Department of Molecular Biology, Medical Biochemistry and Pathology, Laval University, Quebec City, Canada
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, University of Erlangen-Nürnberg, Erlangen, Germany
- Department of Nephrology and Medical Intensive Care, Charite Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Wuttke
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Yurong Cheng
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Tanguy Corre
- Center for Primary Care and Public Health (Unisante), University of Lausanne, Lausanne, Switzerland
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia
| | - Corrinda Black
- Aberdeen Centre for Health Data Science, School of Medicine, Medical Science and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
| | - Vanessa Bruat
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Maria Pina Concas
- Institute for Maternal and Child Health IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo" 34127 Trieste, Italy
| | - Cinzia Sala
- Genetics of Common Disorders Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sven Bergmann
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Department of Integrative Biomedical Sciences, University of Cape Town, Cape Town, South Africa
| | - Harry Campbell
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthias Olden
- Department of Genetic Epidemiology, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Ozren Polasek
- Department of Public Health, Faculty of Medicine, University of Split, Split, Croatia
| | - David J Porteous
- Centre for Genomic & Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Generation Scotland, Centre for Genomic and Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Ian J Deary
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, United Kingdom
| | - Francois Madore
- Division of Nephrology, Hôpital du Sacre-Coeur de Montreal, Montreal, Canada
| | - Philip Awadalla
- Division of Nephrology, Hôpital du Sacre-Coeur de Montreal, Montreal, Canada
| | - Giorgia Girotto
- Institute for Maternal and Child Health IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo" 34127 Trieste, Italy
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34149, Trieste, Italy
| | - Sheila Ulivi
- Institute for Maternal and Child Health IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) "Burlo Garofolo" 34127 Trieste, Italy
| | - David Conen
- Population Health Research Institute, McMaster University, Hamilton, Canada
| | - Elke Wuehl
- Cardiology Division, University Hospital Basel, Basel, Switzerland
| | - Eric Olinger
- Mechanisms of Inherited Kidney Disorders Group, Institute of Physiology Institute of Physiology, University of Zurich, Zurich, Switzerland
- Translational and Clinical Research Institute, Newcastle upon Tyne, Newcastle, United Kingdom
| | - James F Wilson
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
- Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, United Kingdom
| | - Murielle Bochud
- Center for Primary Care and Public Health (Unisante), University of Lausanne, Lausanne, Switzerland
| | - Anna Köttgen
- Institute of Genetic Epidemiology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, University of Edinburgh, Edinburgh, United Kingdom
- Centre for Genomic & Experimental Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Olivier Devuyst
- Mechanisms of Inherited Kidney Disorders Group, Institute of Physiology Institute of Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
89
|
The Interplay between Uremic Toxins and Albumin, Membrane Transporters and Drug Interaction. Toxins (Basel) 2022; 14:toxins14030177. [PMID: 35324674 PMCID: PMC8949274 DOI: 10.3390/toxins14030177] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/21/2022] [Accepted: 02/24/2022] [Indexed: 01/10/2023] Open
Abstract
Uremic toxins are a heterogeneous group of molecules that accumulate in the body due to the progression of chronic kidney disease (CKD). These toxins are associated with kidney dysfunction and the development of comorbidities in patients with CKD, being only partially eliminated by dialysis therapies. Importantly, drugs used in clinical treatments may affect the levels of uremic toxins, their tissue disposition, and even their elimination through the interaction of both with proteins such as albumin and cell membrane transporters. In this context, protein-bound uremic toxins (PBUTs) are highlighted for their high affinity for albumin, the most abundant serum protein with multiple binding sites and an ability to interact with drugs. Membrane transporters mediate the cellular influx and efflux of various uremic toxins, which may also compete with drugs as substrates, and both may alter transporter activity or expression. Therefore, this review explores the interaction mechanisms between uremic toxins and albumin, as well as membrane transporters, considering their potential relationship with drugs used in clinical practice.
Collapse
|
90
|
First genome-wide association study investigating blood pressure and renal traits in domestic cats. Sci Rep 2022; 12:1899. [PMID: 35115544 PMCID: PMC8813908 DOI: 10.1038/s41598-022-05494-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 12/28/2021] [Indexed: 11/08/2022] Open
Abstract
Hypertension (HTN) and chronic kidney disease (CKD) are common in ageing cats. In humans, blood pressure (BP) and renal function are complex heritable traits. We performed the first feline genome-wide association study (GWAS) of quantitative traits systolic BP and creatinine and binary outcomes HTN and CKD, testing 1022 domestic cats with a discovery, replication and meta-analysis design. No variants reached experimental significance level in the discovery stage for any phenotype. Follow up of the top 9 variants for creatinine and 5 for systolic BP, one SNP reached experimental-wide significance for association with creatinine in the combined meta-analysis (chrD1.10258177; P = 1.34 × 10–6). Exploratory genetic risk score (GRS) analyses were performed. Within the discovery sample, GRS of top SNPs from the BP and creatinine GWAS show strong association with HTN and CKD but did not validate in independent replication samples. A GRS including SNPs corresponding to human CKD genes was not significant in an independent subset of cats. Gene-set enrichment and pathway-based analysis (GSEA) was performed for both quantitative phenotypes, with 30 enriched pathways with creatinine. Our results support the utility of GWASs and GSEA for genetic discovery of complex traits in cats, with the caveat of our findings requiring validation.
Collapse
|
91
|
Piras D, Lepori N, Cabiddu G, Pani A. How Genetics Can Improve Clinical Practice in Chronic Kidney Disease: From Bench to Bedside. J Pers Med 2022; 12:jpm12020193. [PMID: 35207681 PMCID: PMC8875178 DOI: 10.3390/jpm12020193] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 01/27/2023] Open
Abstract
Chronic kidney disease (CKD) is considered a major global health problem with high socio-economic costs: the risk of CKD in individuals with an affected first degree relative has been found to be three times higher than in the general population. Genetic factors are known to be involved in CKD pathogenesis, both due to the possible presence of monogenic pathologies as causes of CKD, and to the role of numerous gene variants in determining susceptibility to the development of CKD. The genetic study of CKD patients can represent a useful tool in the hands of the clinician; not only in the diagnostic and prognostic field, but potentially also in guiding therapeutic choices and in designing clinical trials. In this review we discuss the various aspects of the role of genetic analysis on clinical management of patients with CKD with a focus on clinical applications. Several topics are discussed in an effort to provide useful information for daily clinical practice: definition of susceptibility to the development of CKD, identification of unrecognized monogenic diseases, reclassification of the etiological diagnosis, role of pharmacogenetics.
Collapse
Affiliation(s)
- Doloretta Piras
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
- Correspondence:
| | - Nicola Lepori
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
| | - Gianfranca Cabiddu
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, 09134 Cagliari, Italy
| | - Antonello Pani
- Struttura Complessa di Nefrologia, Dialisi e Trapianto, ARNAS Brotzu, 09134 Cagliari, Italy; (N.L.); (G.C.); (A.P.)
- Dipartimento di Scienze Mediche e Sanità Pubblica, Università degli Studi di Cagliari, 09134 Cagliari, Italy
- Istituto di Ricerca Genetica e Biomedica (IRGB), Consiglio Nazionale delle Ricerce (CNR), 09042 Monserrato, Italy
| |
Collapse
|
92
|
Zhang H, Wang A, Xu T, Mo X, Zhang Y. Promoter DNA Methylation in GWAS-Identified Genes as Potential Functional Elements for Blood Pressure: An Observational and Mendelian Randomization Study. Front Genet 2022; 12:791146. [PMID: 35087571 PMCID: PMC8787193 DOI: 10.3389/fgene.2021.791146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 01/03/2023] Open
Abstract
Genome-wide association studies have identified numerous genetic loci for blood pressure (BP). However, the relationships of functional elements inside these loci with BP are not fully understood. This study represented an effort to determine if promoter DNA methylations inside BP-associated loci were associated with BP.We conducted a cross-sectional study investigating the association between promoter DNA methylations of 10 candidate genes and BP in 1,241 Chinese individuals. Twenty-one genomic fragments in the CpG Islands were sequenced. The associations of methylation levels with BP and hypertension were assessed in regression models. Mendelian randomization (MR) analysis was then applied to find supporting evidence for the identified associations.A total of 413 DNA methylation sites were examined in an observational study. Methylation levels of 24 sites in PRDM6, IGFBP3, SYT7, PDE3A, TBX2 and C17orf82 were significantly associated with BP. Methylation levels of PRDM6 and SYT7 were significantly associated with hypertension. Methylation levels of five sites (including cg06713098) in IGFBP3 were significantly associated with DBP. MR analysis found associations between the methylation levels of six CpG sites (cg06713098, cg14228300, cg23193639, cg21268650, cg10677697 and cg04812164) around the IGFBP3 promoter and DBP. Methylation levels of cg14228300 and cg04812164 were associated with SBP. By further applying several MR methods we showed that the associations may not be due to pleiotropy. Association between IGFBP3 mRNA levels in blood cells and BP was also found in MR analysis. This study identified promoter methylation as potential functional element for BP. The identified methylations may be involved in the regulatory pathway linking genetic variants to BP.
Collapse
Affiliation(s)
- Huan Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Aili Wang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Tan Xu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Xingbo Mo
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China.,Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Suzhou, China
| | - Yonghong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Medical College of Soochow University, Suzhou, China.,Department of Epidemiology, School of Public Health, Medical College of Soochow University, Suzhou, China
| |
Collapse
|
93
|
Witasp A, Luttropp K, Qureshi AR, Barany P, Heimbürger O, Wennberg L, Ekström TJ, Shiels PG, Stenvinkel P, Nordfors L. Longitudinal genome-wide DNA methylation changes in response to kidney failure replacement therapy. Sci Rep 2022; 12:470. [PMID: 35013499 PMCID: PMC8748627 DOI: 10.1038/s41598-021-04321-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/13/2021] [Indexed: 01/01/2023] Open
Abstract
Chronic kidney disease (CKD) is an emerging public health priority associated with high mortality rates and demanding treatment regimens, including life-style changes, medications or even dialysis or renal transplantation. Unavoidably, the uremic milieu disturbs homeostatic processes such as DNA methylation and other vital gene regulatory mechanisms. Here, we aimed to investigate how dialysis or kidney transplantation modifies the epigenome-wide methylation signature over 12 months of treatment. We used the Infinium HumanMethylation450 BeadChip on whole blood samples from CKD-patients undergoing either dialysis (n = 11) or kidney transplantation (n = 12) and 24 age- and sex-matched population-based controls. At baseline, comparison between patients and controls identified several significant (PFDR < 0.01) CpG methylation differences in genes with functions relevant to inflammation, cellular ageing and vascular calcification. Following 12 months, the global DNA methylation pattern of patients approached that seen in the control group. Notably, 413 CpG sites remained differentially methylated at follow-up in both treatment groups compared to controls. Together, these data indicate that the uremic milieu drives genome-wide methylation changes that are partially reversed with kidney failure replacement therapy. Differentially methylated CpG sites unaffected by treatment may be of particular interest as they could highlight candidate genes for kidney disease per se.
Collapse
Affiliation(s)
- Anna Witasp
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, M99, 141 86, Stockholm, Sweden
| | - Karin Luttropp
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, M99, 141 86, Stockholm, Sweden
| | - Peter Barany
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, M99, 141 86, Stockholm, Sweden
| | - Olof Heimbürger
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, M99, 141 86, Stockholm, Sweden
| | - Lars Wennberg
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas J Ekström
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Paul G Shiels
- College of Medical, Veterinary and Life Sciences Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Peter Stenvinkel
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, M99, 141 86, Stockholm, Sweden
| | - Louise Nordfors
- Division of Renal Medicine, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Karolinska University Hospital, M99, 141 86, Stockholm, Sweden.
| |
Collapse
|
94
|
Ghasemi S, Becker T, Grabe HJ, Teumer A. Discovery of novel eGFR-associated multiple independent signals using a quasi-adaptive method. Front Genet 2022; 13:997302. [PMID: 36386835 PMCID: PMC9660290 DOI: 10.3389/fgene.2022.997302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 10/13/2022] [Indexed: 11/13/2022] Open
Abstract
A decreased estimated glomerular filtration rate (eGFR) leading to chronic kidney disease is a significant public health problem. Kidney function is a heritable trait, and recent application of genome-wide association studies (GWAS) successfully identified multiple eGFR-associated genetic loci. To increase statistical power for detecting independent associations in GWAS loci, we improved our recently developed quasi-adaptive method estimating SNP-specific alpha levels for the conditional analysis, and applied it to the GWAS meta-analysis results of eGFR among 783,978 European-ancestry individuals. Among known eGFR loci, we revealed 19 new independent association signals that were subsequently replicated in the United Kingdom Biobank (n = 408,608). These associations have remained undetected by conditional analysis using the established conservative genome-wide significance level of 5 × 10-8. Functional characterization of known index SNPs and novel independent signals using colocalization of conditional eGFR association results and gene expression in cis across 51 human tissues identified two potentially causal genes across kidney tissues: TSPAN33 and TFDP2, and three candidate genes across other tissues: SLC22A2, LRP2, and CDKN1C. These colocalizations were not identified in the original GWAS. By applying our improved quasi-adaptive method, we successfully identified additional genetic variants associated with eGFR. Considering these signals in colocalization analyses can increase the precision of revealing potentially functional genes of GWAS loci.
Collapse
Affiliation(s)
- Sahar Ghasemi
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany.,Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| | - Tim Becker
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Hans J Grabe
- Department of Psychiatry and Psychotherapy, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases DZNE, Site Rostock/Greifswald, Greifswald, Germany
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Greifswald, Greifswald, Germany
| |
Collapse
|
95
|
Jiao H, Zhang M, Zhang Y, Wang Y, Li WD. Pathway Association Studies Reveal Gene Loci and Pathway Networks that Associated With Plasma Cystatin C Levels. Front Genet 2021; 12:711155. [PMID: 34899825 PMCID: PMC8656399 DOI: 10.3389/fgene.2021.711155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 11/09/2021] [Indexed: 01/09/2023] Open
Abstract
As a marker for glomerular filtration, plasma cystatin C level is used to evaluate kidney function. To decipher genetic factors that control the plasma cystatin C level, we performed genome-wide association and pathway association studies using United Kingdom Biobank data. One hundred fifteen loci yielded p values less than 1 × 10−100, three genes (clusters) showed the most significant associations, including the CST8-CST9 cluster on chromosome 20, the SH2B3-ATXN2 gene region on chromosome 12, and the SHROOM3-CCDC158 gene region on chromosome 4. In pathway association studies, forty significant pathways had FDR (false discovery rate) and or FWER (family-wise error rate) ≤ 0.001: spermatogenesis, leukocyte trans-endothelial migration, cell adhesion, glycoprotein, membrane lipid, steroid metabolic process, and insulin signaling pathways were among the most significant pathways that associated with the plasma cystatin C levels. We also performed Genome-wide association studies for eGFR, top associated genes were largely overlapped with those for cystatin C.
Collapse
Affiliation(s)
- Hongxiao Jiao
- Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Miaomiao Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yuan Zhang
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,College of Public Health, Tianjin Medical University, Tianjin, China
| | - Yaogang Wang
- College of Public Health, Tianjin Medical University, Tianjin, China
| | - Wei-Dong Li
- Department of Genetics, College of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
96
|
de Carvalho LM, de Sousa GR, Moura R, Saggioro F, Facincani I, Costa R, Kahwage PP, Gomes de Paula Queiroz R, Valera ET, Crovella S, Sandrin-Garcia P. Full-house nephropathy associated with high expression of SPATA5L1 due to a genetic pathogenic variant. Rheumatology (Oxford) 2021; 61:e84-e86. [PMID: 34864870 DOI: 10.1093/rheumatology/keab896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/27/2021] [Accepted: 11/20/2021] [Indexed: 11/14/2022] Open
Affiliation(s)
- Luciana Martins de Carvalho
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Division of Pediatric Rheumatology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Ronald Moura
- Department of Advanced Diagnostics, Institute for Maternal and Child Health-IRCCS "Burlo Garofolo", Trieste, Italy
| | - Fabiano Saggioro
- Department of Pathology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Department of Pathology, Rede D'Or-São Paulo (SP), Brazil
| | - Inalda Facincani
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Roberto Costa
- Department of Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Paola Pinheiro Kahwage
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,Division of Pediatric Rheumatology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sergio Crovella
- Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar, P.O. Box: 2713
| | - Paula Sandrin-Garcia
- Keizo Asami Immunopathology Laboratory, Federal University of Pernambuco, Recife, Brazil.,Genetics Department, Federal University of Pernambuco, Recife, Brazil
| |
Collapse
|
97
|
Abstract
Over the past decade, substantial progress has been made in the discovery of alleles contributing to the risk of coronary artery disease. In addition to providing causal insights into disease, these endeavours have yielded and enabled the refinement of polygenic risk scores. These scores can be used to predict incident coronary artery disease in multiple cohorts and indicate the clinical response to some preventive therapies in post hoc analyses of clinical trials. These observations and the widespread ability to calculate polygenic risk scores from direct-to-consumer and health-care-associated biobanks have raised many questions about responsible clinical adoption. In this Review, we describe technical and downstream considerations for the derivation and validation of polygenic risk scores and current evidence for their efficacy and safety. We discuss the implementation of these scores in clinical medicine for uses including risk prediction and screening algorithms for coronary artery disease, prioritization of patient subgroups that are likely to derive benefit from treatment, and efficient prospective clinical trial designs.
Collapse
|
98
|
Lecamwasam A, Novakovic B, Meyer B, Ekinci EI, Dwyer KM, Saffery R. DNA methylation profiling identifies epigenetic differences between early versus late stages of diabetic chronic kidney disease. Nephrol Dial Transplant 2021; 36:2027-2038. [PMID: 33146725 DOI: 10.1093/ndt/gfaa226] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND We investigated a cross-sectional epigenome-wide association study of patients with early and late diabetes-associated chronic kidney disease (CKD) to identify possible epigenetic differences between the two groups as well as changes in methylation across all stages of diabetic CKD. We also evaluated the potential of using a panel of identified 5'-C-phosphate-G-3' (CpG) sites from this cohort to predict the progression of diabetic CKD. METHODS This cross-sectional study recruited 119 adults. DNA was extracted from blood using the Qiagen QIAampDNA Mini Spin Kit. Genome-wide methylation analysis was performed using Illumina Infinium MethylationEPIC BeadChips (HM850K). Intensity data files were processed and analysed using the minfi and MissMethyl packages for R. We examined the degree of methylation of CpG sites in early versus late diabetic CKD patients for CpG sites with an unadjusted P-value <0.01 and an absolute change in methylation of 5% (n = 239 CpG sites). RESULTS Hierarchical clustering of the 239 CpG sites largely separated the two groups. A heat map for all 239 CpG sites demonstrated distinct methylation patterns in the early versus late groups, with CpG sites showing evidence of progressive change. Based on our differentially methylated region (DMR) analysis of the 239 CpG sites, we highlighted two DMRs, namely the cysteine-rich secretory protein 2 (CRISP2) and piwi-like RNA-mediated gene silencing 1 (PIWIL1) genes. The best predictability for the two groups involved a receiver operating characteristics curve of eight CpG sites alone and achieved an area under the curve of 0.976. CONCLUSIONS We have identified distinct DNA methylation patterns between early and late diabetic CKD patients as well as demonstrated novel findings of potential progressive methylation changes across all stages (1-5) of diabetic CKD at specific CpG sites. We have also identified associated genes CRISP2 and PIWIL1, which may have the potential to act as stage-specific diabetes-associated CKD markers, and showed that the use of a panel of eight identified CpG sites alone helps to increase the predictability for the two groups.
Collapse
Affiliation(s)
| | - Boris Novakovic
- Epigenetics Research, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Braydon Meyer
- Epigenetics Research, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Elif I Ekinci
- Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Melbourne, Victoria, Australia
| | - Karen M Dwyer
- School of Medicine, Faculty of Health, Deakin University, Melbourne, Victoria, Australia
| | - Richard Saffery
- Epigenetics Research, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
99
|
Govender MA, Brandenburg JT, Fabian J, Ramsay M. The Use of 'Omics for Diagnosing and Predicting Progression of Chronic Kidney Disease: A Scoping Review. Front Genet 2021; 12:682929. [PMID: 34819944 PMCID: PMC8606569 DOI: 10.3389/fgene.2021.682929] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 10/18/2021] [Indexed: 12/19/2022] Open
Abstract
Globally, chronic kidney disease (CKD) contributes substantial morbidity and mortality. Recently, various 'omics platforms have provided insight into the molecular basis of kidney dysfunction. This scoping review is a synthesis of the current literature on the use of different 'omics platforms to identify biomarkers that could be used to detect early-stage CKD, predict disease progression, and identify pathways leading to CKD. This review includes 123 articles published from January 2007 to May 2021, following a structured selection process. The most common type of 'omic platform was proteomics, appearing in 55 of the studies and two of these included a metabolomics component. Most studies (n = 91) reported on CKD associated with diabetes mellitus. Thirteen studies that provided information on the biomarkers associated with CKD and explored potential pathways involved in CKD are discussed. The biomarkers that are associated with risk or early detection of CKD are SNPs in the MYH9/APOL1 and UMOD genes, the proteomic CKD273 biomarker panel and metabolite pantothenic acid. Pantothenic acid and the CKD273 biomarker panel were also involved in predicting CKD progression. Retinoic acid pathway genes, UMOD, and pantothenic acid provided insight into potential pathways leading to CKD. The biomarkers were mainly used to detect CKD and predict progression in high-income, European ancestry populations, highlighting the need for representative 'omics research in other populations with disparate socio-economic strata, including Africans, since disease etiologies may differ across ethnic groups. To assess the transferability of findings, it is essential to do research in diverse populations.
Collapse
Affiliation(s)
- Melanie A. Govender
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Jean-Tristan Brandenburg
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - June Fabian
- Wits Donald Gordon Medical Centre, School of Clinical Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Michèle Ramsay
- Division of Human Genetics, National Health Laboratory Service and School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Sydney Brenner Institute for Molecular Bioscience, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
100
|
Ding Y, Xu Z, Pan Y, Meng X, Xiang X, Li H, Liu L, Wang Y. Association Between CST3 Gene Polymorphisms and Large-Artery Atherosclerotic Stroke. Front Neurol 2021; 12:738148. [PMID: 34721268 PMCID: PMC8548665 DOI: 10.3389/fneur.2021.738148] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/07/2021] [Indexed: 11/25/2022] Open
Abstract
Objective: Cystatin C, a marker of atherosclerosis, is encoded by CST3. We aimed to evaluate whether two single-nucleotide polymorphisms (SNPs) of CST3 are correlated with large-artery atherosclerotic stroke (LAAS) and prognosis. Methods: This subgroup analysis of the Third China National Stroke Registry (CNSR-III) enrolled acute ischemic stroke (AIS) patients within 7 days from August 2015 to March 2018 in China. rs13038305 and rs911119 of CST3 were selected based on the strong association with cystatin C concentration. Results: Two loci of CST3 (rs13038305 and rs911119) were analyzed in 3,833 ischemic stroke patients. Carriers of T allele in rs13038305 and C allele in rs911119 tend to have lower serum cystatin C levels (p < 0.05). Compared with C/C as a reference in rs13038305, odds ratio (OR) of T/T was 0.486, 95% CI 0.237–0.994, p = 0.048. Per C allele of rs13038305 also showed an increased level of low-density lipoprotein cholesterol (LDL-C), β (95% CI) was 1.335 (1.008–1.250), p = 0.044. No correlation was found between the selected SNPs and stroke prognosis (functional outcome, recurrence, and mortality). Conclusions: Carriers of the T allele in rs13038305 tend to have a lower proportion of LAAS. rs13038305 and rs911119 polymorphisms were likely to affect cystatin C concentration independently of kidney function.
Collapse
Affiliation(s)
- Yarong Ding
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Zhe Xu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yuesong Pan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Xia Meng
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Xianglong Xiang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Hao Li
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Liping Liu
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| | - Yongjun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Center of Stroke, Beijing Institute for Brain Disorders, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China
| |
Collapse
|