J Pediatr Genet 2016; 05(01): 069-075
DOI: 10.1055/s-0035-1557113
Review Article
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

Actualizing the Benefits of Genomic Discovery in Pediatric Nephrology

Matthew G. Sampson
1   Division of Pediatric Nephrology, Department of Pediatrics and Communicable Diseases, University of Michigan School of Medicine, Ann Arbor, Michigan, United States
› Author Affiliations
Further Information

Publication History

20 December 2014

19 January 2015

Publication Date:
13 August 2015 (online)

Abstract

The discovery of genetic variation associated with pediatric kidney disease has shed light on the biology underlying these conditions and, in some cases, has improved our clinical management of patients. We are challenged to continue the momentum of the genomic era in pediatric nephrology by identifying novel disease-associated genetic variation and translating these discoveries into clinical applications. This article reviews the diverse forms of genetic architecture that have been found to be associated with kidney diseases and traits. These include rare, fully penetrant variants responsible for Mendelian forms of disease, copy number variants, and more common variants associated with increased risk of disease. These discoveries have provided us with a greater understanding of the molecular mechanisms underlying these conditions and highlighted key pathways for potential intervention. In a number of areas, the identification of rare, fully penetrant variants is immediately clinically relevant, whether in regard to diagnostic testing, prediction of outcomes, or choice of therapies and interventions. This article discusses limitations in the deterministic view of rare, putatively causal mutations, a challenge increasing in importance as sequencing expands to many more genes and patients. This article also focusses on common genetic variants, using those found to be associated with focal segmental glomerulosclerosis in African-Americans, IgA nephropathy, chronic kidney disease (CKD), and estimated glomerular filtration rate (eGFR) as examples. Identifying common genetic variants associated with disease will complement other areas of genomic inquiry, lead to a greater biological understanding of disease, and will benefit pediatric nephrology patients.

 
  • References

  • 1 Toward Precision Medicine: Building a Knowledge Network for Biomedical Research and a New Taxonomy of Disease. Washington, DC; 2011
  • 2 Schrodi SJ, Mukherjee S, Shan Y , et al. Genetic-based prediction of disease traits: prediction is very difficult, especially about the future. Front Genet 2014; 5: 162
  • 3 Brophy PD, Alasti F, Darbro BW , et al. Genome-wide copy number variation analysis of a Branchio-oto-renal syndrome cohort identifies a recombination hotspot and implicates new candidate genes. Hum Genet 2013; 132 (12) 1339-1350
  • 4 Sanna-Cherchi S, Kiryluk K, Burgess KE , et al. Copy-number disorders are a common cause of congenital kidney malformations. Am J Hum Genet 2012; 91 (6) 987-997
  • 5 Stanescu HC, Arcos-Burgos M, Medlar A , et al. Risk HLA-DQA1 and PLA(2)R1 alleles in idiopathic membranous nephropathy. N Engl J Med 2011; 364 (7) 616-626
  • 6 Genovese G, Tonna SJ, Knob AU , et al. A risk allele for focal segmental glomerulosclerosis in African Americans is located within a region containing APOL1 and MYH9. Kidney Int 2010; 78 (7) 698-704
  • 7 Kopp JB, Nelson GW, Sampath K , et al. APOL1 genetic variants in focal segmental glomerulosclerosis and HIV-associated nephropathy. J Am Soc Nephrol 2011; 22 (11) 2129-2137
  • 8 Gbadegesin RA, Adeyemo A, Webb NJ , et al; Members of the Mid-West Pediatric Nephrology Consortium. HLA-DQA1 and PLCG2 Are Candidate Risk Loci for Childhood-Onset Steroid-Sensitive Nephrotic Syndrome. J Am Soc Nephrol 2014;
  • 9 Okamoto K, Tokunaga K, Doi K , et al. Common variation in GPC5 is associated with acquired nephrotic syndrome. Nat Genet 2011; 43 (5) 459-463
  • 10 Gharavi AG, Kiryluk K, Choi M , et al. Genome-wide association study identifies susceptibility loci for IgA nephropathy. Nat Genet 2011; 43 (4) 321-327
  • 11 Kiryluk K, Li Y, Scolari F , et al. Discovery of new risk loci for IgA nephropathy implicates genes involved in immunity against intestinal pathogens. Nat Genet 2014; 46 (11) 1187-1196
  • 12 Murray B, Hawes E, Lee RA, Watson R, Roederer MW. Genes and beans: pharmacogenomics of renal transplant. Pharmacogenomics 2013; 14 (7) 783-798
  • 13 Lieske JC, Monico CG, Holmes WS , et al. International registry for primary hyperoxaluria. Am J Nephrol 2005; 25 (3) 290-296
  • 14 Monico CG, Rossetti S, Olson JB, Milliner DS. Pyridoxine effect in type I primary hyperoxaluria is associated with the most common mutant allele. Kidney Int 2005; 67 (5) 1704-1709
  • 15 Lemaire M, Frémeaux-Bacchi V, Schaefer F , et al. Recessive mutations in DGKE cause atypical hemolytic-uremic syndrome. Nat Genet 2013; 45 (5) 531-536
  • 16 Westland R, Bodria M, Carrea A , et al. Phenotypic expansion of DGKE-associated diseases. J Am Soc Nephrol 2014; 25 (7) 1408-1414
  • 17 Fivush BA, Jabs K, Neu AM , et al. Chronic renal insufficiency in children and adolescents: the 1996 annual report of NAPRTCS. North American Pediatric Renal Transplant Cooperative Study. Pediatr Nephrol 1998; 12 (4) 328-337
  • 18 Weber S. Novel genetic aspects of congenital anomalies of kidney and urinary tract. Curr Opin Pediatr 2012; 24 (2) 212-218
  • 19 Kohl S, Hwang DY, Dworschak GC , et al. Mild recessive mutations in six Fraser syndrome-related genes cause isolated congenital anomalies of the kidney and urinary tract. J Am Soc Nephrol 2014; 25 (9) 1917-1922
  • 20 Hwang DY, Dworschak GC, Kohl S , et al. Mutations in 12 known dominant disease-causing genes clarify many congenital anomalies of the kidney and urinary tract. Kidney Int 2014; 85 (6) 1429-1433
  • 21 Lipska BS, Ranchin B, Iatropoulos P , et al; PodoNet Consortium. Genotype-phenotype associations in WT1 glomerulopathy. Kidney Int 2014; 85 (5) 1169-1178
  • 22 Chernin G, Vega-Warner V, Schoeb DS , et al; Members of the GPN Study Group. Genotype/phenotype correlation in nephrotic syndrome caused by WT1 mutations. Clin J Am Soc Nephrol 2010; 5 (9) 1655-1662
  • 23 Ruf RG, Lichtenberger A, Karle SM , et al; Arbeitsgemeinschaft Für Pädiatrische Nephrologie Study Group. Patients with mutations in NPHS2 (podocin) do not respond to standard steroid treatment of nephrotic syndrome. J Am Soc Nephrol 2004; 15 (3) 722-732
  • 24 Maas RJ, Deegens JK, van den Brand JA, Cornelissen EA, Wetzels JF. A retrospective study of focal segmental glomerulosclerosis: clinical criteria can identify patients at high risk for recurrent disease after first renal transplantation. BMC Nephrol 2013; 14: 47
  • 25 Rodríguez de Córdoba S, Hidalgo MS, Pinto S, Tortajada A. Genetics of atypical hemolytic uremic syndrome (aHUS). Semin Thromb Hemost 2014; 40 (4) 422-430
  • 26 Kavanagh D, Richards A, Goodship T, Jalanko H. Transplantation in atypical hemolytic uremic syndrome. Semin Thromb Hemost 2010; 36 (6) 653-659
  • 27 Barua M, Brown EJ, Charoonratana VT, Genovese G, Sun H, Pollak MR. Mutations in the INF2 gene account for a significant proportion of familial but not sporadic focal and segmental glomerulosclerosis. Kidney Int 2013; 83 (2) 316-322
  • 28 Gbadegesin RA, Lavin PJ, Hall G , et al. Inverted formin 2 mutations with variable expression in patients with sporadic and hereditary focal and segmental glomerulosclerosis. Kidney Int 2012; 81 (1) 94-99
  • 29 Saleem MA. New developments in steroid-resistant nephrotic syndrome. Pediatr Nephrol 2013; 28 (5) 699-709
  • 30 Benzing T, Schermer B. Clinical spectrum and pathogenesis of nephronophthisis. Curr Opin Nephrol Hypertens 2012; 21 (3) 272-278
  • 31 Vehaskari VM. Heritable forms of hypertension. Pediatr Nephrol 2009; 24 (10) 1929-1937
  • 32 Halbritter J, Diaz K, Chaki M , et al. High-throughput mutation analysis in patients with a nephronophthisis-associated ciliopathy applying multiplexed barcoded array-based PCR amplification and next-generation sequencing. J Med Genet 2012; 49 (12) 756-767
  • 33 Lovric S, Fang H, Vega-Warner V , et al; Nephrotic Syndrome Study Group. Rapid detection of monogenic causes of childhood-onset steroid-resistant nephrotic syndrome. Clin J Am Soc Nephrol 2014; 9 (6) 1109-1116
  • 34 Halbritter J, Baum M, Hynes AM , et al. Fourteen Monogenic Genes Account for 15% of Nephrolithiasis/Nephrocalcinosis. J Am Soc Nephrol 2015; 26 (3) 543-551
  • 35 Chatterjee R, Hoffman M, Cliften P, Seshan S, Liapis H, Jain S. Targeted exome sequencing integrated with clinicopathological information reveals novel and rare mutations in atypical, suspected and unknown cases of Alport syndrome or proteinuria. PLoS ONE 2013; 8 (10) e76360
  • 36 Sadowski CE, Lovric S, Ashraf S , et al; the SRNS Study Group. A single-gene cause in 29.5% of cases of steroid-resistant nephrotic syndrome. J Am Soc Nephrol 2015; 26: 1279-1289
  • 37 Rood IM, Deegens JK, Wetzels JF. Genetic causes of focal segmental glomerulosclerosis: implications for clinical practice. Nephrol Dial Transplant 2012; 27 (3) 882-890
  • 38 Tennessen JA, Bigham AW, O'Connor TD , et al; Broad GO; Seattle GO; NHLBI Exome Sequencing Project. Evolution and functional impact of rare coding variation from deep sequencing of human exomes. Science 2012; 337 (6090) 64-69
  • 39 Tennessen JA, O'Connor TD, Bamshad MJ, Akey JM. The promise and limitations of population exomics for human evolution studies. Genome Biol 2011; 12 (9) 127
  • 40 Roberts IS. Pathology of IgA nephropathy. Nat Rev Nephrol 2014; 10 (8) 445-454
  • 41 Wyatt RJ, Julian BA. IgA nephropathy. N Engl J Med 2013; 368 (25) 2402-2414
  • 42 Novak J, Julian BA, Mestecky J, Renfrow MB. Glycosylation of IgA1 and pathogenesis of IgA nephropathy. Semin Immunopathol 2012; 34 (3) 365-382
  • 43 Köttgen A, Pattaro C, Böger CA , et al. New loci associated with kidney function and chronic kidney disease. Nat Genet 2010; 42 (5) 376-384
  • 44 Friedman DJ, Kozlitina J, Genovese G, Jog P, Pollak MR. Population-based risk assessment of APOL1 on renal disease. J Am Soc Nephrol 2011; 22 (11) 2098-2105
  • 45 Friedman DJ, Pollak MR. Genetics of kidney failure and the evolving story of APOL1. J Clin Invest 2011; 121 (9) 3367-3374
  • 46 Pollak MR, Genovese G, Friedman DJ. APOL1 and kidney disease. Curr Opin Nephrol Hypertens 2012; 21 (2) 179-182
  • 47 O'Seaghdha CM, Yang Q, Wu H, Hwang SJ, Fox CS. Performance of a genetic risk score for CKD stage 3 in the general population. Am J Kidney Dis 2012; 59 (1) 19-24
  • 48 Paynter NP, Chasman DI, Paré G , et al. Association between a literature-based genetic risk score and cardiovascular events in women. JAMA 2010; 303 (7) 631-637
  • 49 Reeves-Daniel AM, DePalma JA, Bleyer AJ , et al. The APOL1 gene and allograft survival after kidney transplantation. Am J Transplant 2011; 11 (5) 1025-1030
  • 50 Lee BT, Kumar V, Williams TA , et al. The APOL1 genotype of African American kidney transplant recipients does not impact 5-year allograft survival. Am J Transplant 2012; 12 (7) 1924-1928
  • 51 Kao WH, Klag MJ, Meoni LA , et al; Family Investigation of Nephropathy and Diabetes Research Group. MYH9 is associated with nondiabetic end-stage renal disease in African Americans. Nat Genet 2008; 40 (10) 1185-1192
  • 52 Ito K, Bick AG, Flannick J , et al. Increased burden of cardiovascular disease in carriers of APOL1 genetic variants. Circ Res 2014; 114 (5) 845-850
  • 53 Parsa A, Kao WH, Xie D , et al; AASK Study Investigators; CRIC Study Investigators. APOL1 risk variants, race, and progression of chronic kidney disease. N Engl J Med 2013; 369 (23) 2183-2196
  • 54 Tzur S, Rosset S, Skorecki K, Wasser WG. APOL1 allelic variants are associated with lower age of dialysis initiation and thereby increased dialysis vintage in African and Hispanic Americans with non-diabetic end-stage kidney disease. Nephrol Dial Transplant 2012; 27 (4) 1498-1505
  • 55 Coenen MJ, Hofstra JM, Debiec H , et al. Phospholipase A2 receptor (PLA2R1) sequence variants in idiopathic membranous nephropathy. J Am Soc Nephrol 2013; 24 (4) 677-683
  • 56 Lv J, Hou W, Zhou X , et al. Interaction between PLA2R1 and HLA-DQA1 variants associates with anti-PLA2R antibodies and membranous nephropathy. J Am Soc Nephrol 2013; 24 (8) 1323-1329
  • 57 Gigarel N, Frydman N, Burlet P , et al. Preimplantation genetic diagnosis for autosomal recessive polycystic kidney disease. Reprod Biomed Online 2008; 16 (1) 152-158
  • 58 Rood IM, Deegens JK, Merchant ML , et al. Comparison of three methods for isolation of urinary microvesicles to identify biomarkers of nephrotic syndrome. Kidney Int 2010; 78 (8) 810-816
  • 59 Büscher AK, Kranz B, Büscher R , et al. Immunosuppression and renal outcome in congenital and pediatric steroid-resistant nephrotic syndrome. Clin J Am Soc Nephrol 2010; 5 (11) 2075-2084
  • 60 Weber S, Gribouval O, Esquivel EL , et al. NPHS2 mutation analysis shows genetic heterogeneity of steroid-resistant nephrotic syndrome and low post-transplant recurrence. Kidney Int 2004; 66 (2) 571-579
  • 61 Ovunc B, Ashraf S, Vega-Warner V , et al; Gesellschaft für Pädiatrische Nephrologie (GPN) Study Group. Mutation analysis of NPHS1 in a worldwide cohort of congenital nephrotic syndrome patients. Nephron Clin Pract 2012; 120 (3) c139-c146
  • 62 Heeringa SF, Vlangos CN, Chernin G , et al; Members of the APN Study Group. Thirteen novel NPHS1 mutations in a large cohort of children with congenital nephrotic syndrome. Nephrol Dial Transplant 2008; 23 (11) 3527-3533
  • 63 Wong W, Morris MC, Kara T. Congenital nephrotic syndrome with prolonged renal survival without renal replacement therapy. Pediatr Nephrol 2013; 28 (12) 2313-2321
  • 64 Matejas V, Hinkes B, Alkandari F , et al. Mutations in the human laminin beta2 (LAMB2) gene and the associated phenotypic spectrum. Hum Mutat 2010; 31 (9) 992-1002
  • 65 Hinkes B, Wiggins RC, Gbadegesin R , et al. Positional cloning uncovers mutations in PLCE1 responsible for a nephrotic syndrome variant that may be reversible. Nat Genet 2006; 38 (12) 1397-1405
  • 66 Weeke P, Denny JC, Basterache L , et al. Examining Rare and Low-Frequency Genetic Variants Previously Associated with Lone or Familial Forms of Atrial Fibrillation in an Electronic Medical Record System: A Cautionary Note. Circ Cardiovasc Genet 2015; 8 (1) 58-63