Clinical utility of genetic testing in early-onset kidney disease: seven genes are the main players

Author(s):  
Andrea Domingo-Gallego ◽  
Marc Pybus ◽  
Gemma Bullich ◽  
Mónica Furlano ◽  
Laia Ejarque-Vila ◽  
...  

Abstract Background Inherited kidney diseases are one of the leading causes of chronic kidney disease (CKD) that manifests before the age of 30 years. Precise clinical diagnosis of early-onset CKD is complicated due to the high phenotypic overlap, but genetic testing is a powerful diagnostic tool. We aimed to develop a genetic testing strategy to maximize the diagnostic yield for patients presenting with early-onset CKD and to determine the prevalence of the main causative genes. Methods We performed genetic testing of 460 patients with early-onset CKD of suspected monogenic cause using next-generation sequencing of a custom-designed kidney disease gene panel in addition to targeted screening for c.428dupC MUC1. Results We achieved a global diagnostic yield of 65% (300/460), which varied depending on the clinical diagnostic group: 77% in cystic kidney diseases, 76% in tubulopathies, 67% in autosomal dominant tubulointerstitial kidney disease, 61% in glomerulopathies, and 38% in congenital anomalies of the kidney and urinary tract. Among the 300 genetically diagnosed patients, the clinical diagnosis was confirmed in 77%, a specific diagnosis within a clinical diagnostic group was identified in 15%, and 7% of cases were reclassified. Of the 64 causative genes identified in our cohort, seven (COL4A3, COL4A4, COL4A5, HNF1B, PKD1, PKD2, and PKHD1) accounted for 66% (198/300) of the genetically diagnosed patients. Conclusions Two-thirds of patients with early-onset CKD in this cohort had a genetic cause. Just seven genes were responsible for the majority of diagnoses. Establishing a genetic diagnosis is crucial to define the precise etiology of CKD, which allows accurate genetic counseling and improved patient management.

Kidney360 ◽  
2020 ◽  
pp. 10.34067/KID.0002272020
Author(s):  
Nasim Bekheirnia ◽  
Kevin E. Glinton ◽  
Linda Rossetti ◽  
Joshua Manor ◽  
Wuyan Chen ◽  
...  

Background: As genetic testing increasingly integrates into the practice of nephrology, our understanding of the basis of many kidney disorders has exponentially increased. Given this, we recently initiated a Renal Genetics Clinic (RGC) at our large, urban children's hospital for patients with kidney disorders. Methods: Genetic testing was performed in Clinical Laboratory Improvement Amendments (CLIA) certified laboratories using single gene testing, multi-gene panels, chromosomal microarray (CMA), or exome sequencing (ES). Results: A total of 192 patients were evaluated in this clinic, with cystic kidney disease (49/192) being the most common reason for referral followed by Congenital Anomalies of the Kidney and Urinary Tract (CAKUT: 41/192) and hematuria (38/192). Genetic testing was performed for 158 patients with an overall diagnostic yield of 81/158 (51.3%). In the patients who reached a genetic diagnosis, 16/81 (19.7%), medical or surgical treatment of the patients were impacted, and in 12/81 (14.8%), previous clinical diagnoses were changed to more accurate genetic diagnoses. Conclusions: Such testing provided an accurate diagnosis for children and in some cases led to further diagnosis in seemingly asymptomatic family members and changes to overall medical management. Genetic testing, as facilitated by such a specialized clinical setting, thus appears to have clear utility in the diagnosis and counseling of patients with a wide range of kidney manifestations.


With the advancement in, and increased availability of, genetic testing and a rapidly growing understanding of the genetic basis of many kidney diseases, it is important that nephrologists understand the rationale, limitations, and implications of making a genetic diagnosis in patients with kidney disease. While most inherited kidney diseases are rare, all nephrologists will encounter patients with genetic forms of kidney disease, particularly the cystic kidney diseases. An understanding of the genotype–phenotype relationships across the different inherited kidney diseases provides the nephrologist with an unprecedented understanding of disease pathogenesis. The evolving use of gene-based and molecular therapies utilizing silencing RNA molecules in kidney diseases are likely to become a growing therapeutic option in the future. Nephrologists will need to understand and embrace these new therapies, particularly for this group of kidney diseases.


Author(s):  
M Adela Mansilla ◽  
Ramakrishna R Sompallae ◽  
Carla J Nishimura ◽  
Anne E Kwitek ◽  
Mycah J Kimble ◽  
...  

Abstract Background The clinical diagnosis of genetic renal diseases may be limited by the overlapping spectrum of manifestations between diseases or by the advancement of disease where clues to the original process are absent. The objective of this study was to determine whether genetic testing informs diagnosis and facilitates management of kidney disease patients. Methods We developed a comprehensive genetic testing panel (KidneySeq) to evaluate patients with various phenotypes including cystic diseases, congenital anomalies of the kidney and urinary tract (CAKUT), tubulointerstitial diseases, transport disorders and glomerular diseases. We evaluated this panel in 127 consecutive patients ranging in age from newborns to 81 years who had samples sent in for genetic testing. Results The performance of the sequencing pipeline for single-nucleotide variants was validated using CEPH (Centre de’Etude du Polymorphism) controls and for indels using Genome-in-a-Bottle. To test the reliability of the copy number variant (CNV) analysis, positive samples were re-sequenced and analyzed. For patient samples, a multidisciplinary review board interpreted genetic results in the context of clinical data. A genetic diagnosis was made in 54 (43%) patients and ranged from 54% for CAKUT, 53% for ciliopathies/tubulointerstitial diseases, 45% for transport disorders to 33% for glomerulopathies. Pathogenic and likely pathogenic variants included 46% missense, 11% nonsense, 6% splice site variants, 23% insertion–deletions and 14% CNVs. In 13 cases, the genetic result changed the clinical diagnosis. Conclusion Broad genetic testing should be considered in the evaluation of renal patients as it complements other tests and provides insight into the underlying disease and its management.


2021 ◽  
Vol 36 (Supplement_1) ◽  
Author(s):  
Francesca Becherucci ◽  
Viviana Palazzo ◽  
Luigi Cirillo ◽  
Benedetta Mazzinghi ◽  
Samuela Landini ◽  
...  

Abstract Background and Aims Bartter (BS) and Gitelman syndrome (GS) are autosomal recessive rare inherited disorders characterized by hypokalemic metabolic alkalosis and secondary hyperaldosteronism. The primary defect is a genetically determined impairment of sodium chloride reabsorption in the renal tubule, thus resulting in salt loss, dehydration and acid-base homeostasis perturbations. Although the diagnosis can be suspected based on presenting features, the clinical diagnosis of BS and GS can be challenging, as they are rare and phenotypically overlapping. As a consequence, the current clinical classification lacks of specificity and genetic testing represents the gold standard for the diagnosis. Driven by the rapidly decreasing costs and turn-around time, next-generation sequencing technologies are increasingly utilized in diagnostics and research of inherited tubulopathies, including BS and GS. Recently, sequencing of selected gene panels provided the advantage of achieving high coverage of genes of interest at lower costs, providing high diagnostic yield and new insights into the phenotypic spectrum of these rare disorders. However, whole-exome (WES) is not routinely performed for the molecular diagnosis of BS and GS. The aim of our study was to assess the diagnostic performance of WES in BS and GS and to establish genotype-phenotype correlations. Method We performed WES in all consecutive patients referred for genetic testing with a clinical suspect of BS or GS. Variant prioritization was carried out according to the American College of Medical Genetics and Genomics guidelines (ACMG). Parents and first-degree relatives were included, whenever available. Demographic, clinical and laboratory data were collected retrospectively, in order to establish genotype-phenotype correlations. Results We enrolled 50 patients (22 males, 46 Caucasians) with a clinical diagnosis of BS (19), GS (24) or BS/GS (7). All the patients showed hypokalemic metabolic alkalosis at onset (serum bicarbonate=29.5 mEq/l ± 4.4, potassium= 2.7 mEq/l ± 0.6). The median age at clinical diagnosis was 7 years (range 0-67 years). Three patients had familial history of tubulopathies. WES showed pathogenic variants in 42/50 patients (84%), thus establishing a conclusive diagnosis. Interestingly, a dedicated analytic pipeline allowed us to identify copy number variations (CNVs) in 7/42 patients with a confirmed genetic diagnosis. In detail, WES allowed us to confirm the clinical diagnosis in 33/50 patients, with an improvement in classification in at least 14 cases (i.e. subtype I-V of BS). In 9 additional patients, genetic testing changed the clinical diagnosis: 6 patients with a clinical of BS turned out to have pathogenic variants in SLC12A3, resulting in GS; in 3 patients, genetic testing revised the clinical diagnosis indicating inherited disorders outside the BS/GS spectrum (HELIX syndrome, Primary familial hypoparatiroidism, Type 2 renal hypomagnesemia). Only 38% of patients with a genetic diagnosis of BS showed nephrocalcinosis. Strikingly, this was present in 8% of patients with GS. On the other hand, hypomagnesemia, a distinctive feature of GS, was similarly distributed among BS and GS patients (45% vs. 68%, respectively). Finally, although patients with GS showed a median age at onset higher than patients with BS, some overlap did exist, making differential diagnosis challenging at single-patient level. Conclusion The results of our study demonstrate that WES ensures a high diagnostic yield (84%) in patients with a clinical diagnosis of BS or GS, especially if coupled with analysis of CNVs. This approach showed to be useful in dealing with the phenotypic heterogeneity typical of these rare disorders, improving differential diagnosis by detecting phenocopies also outside the BS/GS spectrum, enabling additional specific work-up, genetic counseling, and screening of at-risk relatives.


Nephron ◽  
2021 ◽  
pp. 1-12
Author(s):  
Kirsty M. Rooney ◽  
Adrian S. Woolf ◽  
Susan J. Kimber

<b><i>Background:</i></b> Kidney disease causes major suffering and premature mortality worldwide. With no cure for kidney failure currently available, and with limited options for treatment, there is an urgent need to develop effective pharmaceutical interventions to slow or prevent kidney disease progression. <b><i>Summary:</i></b> In this review, we consider the feasibility of using human pluripotent stem cell-derived kidney tissues, or organoids, to model genetic kidney disease. Notable successes have been made in modelling genetic tubular diseases (e.g., cystinosis), polycystic kidney disease, and medullary cystic kidney disease. Organoid models have also been used to test novel therapies that ameliorate aberrant cell biology. Some progress has been made in modelling congenital glomerular disease, even though glomeruli within organoids are developmentally immature. Less progress has been made in modelling structural kidney malformations, perhaps because sufficiently mature metanephric mesenchyme-derived nephrons, ureteric bud-derived branching collecting ducts, and a prominent stromal cell population are not generated together within a single protocol. <b><i>Key Messages:</i></b> We predict that the field will advance significantly if organoids can be generated with a full complement of cell lineages and with kidney components displaying key physiological functions, such as glomerular filtration. The future economic upscaling of reproducible organoid generation will facilitate more widespread research applications, including the potential therapeutic application of these stem cell-based technologies.


Author(s):  
Tomáš Seeman ◽  
Magdaléna Fořtová ◽  
Bruno Sopko ◽  
Richard Průša ◽  
Michael Pohl ◽  
...  

Background Hypomagnesaemia is present in 40–50% of children with autosomal dominant renal cysts and diabetes syndrome (RCAD). On the contrary, the prevalence of hypomagnesaemia in children with autosomal dominant polycystic kidney disease (ADPKD) has never been examined. We aimed to investigate whether hypomagnesaemia is present in children with polycystic kidney diseases. Methods Children with cystic kidney diseases were investigated in a cross-sectional study. Serum concentrations of magnesium (S-Mg) and fractional excretion of magnesium (FE-Mg) were tested. Fifty-four children with ADPKD ( n = 26), autosomal recessive polycystic kidney disease (ARPKD) ( n = 16) and RCAD ( n = 12) with median age of 11.2 (0.6–18.6) years were investigated. Results Hypomagnesaemia (S-Mg < 0.7 mmol/L) was detected in none of the children with ADPKD/ARPKD and in eight children (67%) with RCAD. Median S-Mg in children with ADPKD/ARPKD was significantly higher than in children with RCAD (0.89 vs. 0.65 mmol/L, P < 0.01). The FE-Mg was increased in 23% of patients with ADPKD/ARPKD (all had chronic kidney disease stages 2–4) and in 63% of patients with RCAD, where it significantly correlated with estimated glomerular filtration rate (r = −0.87, P < 0.01). Conclusions Hypomagnesaemia is absent in children with ADPKD or ARPKD and could serve as a marker for differential diagnostics between ADPKD, ARPKD and RCAD in children with cystic kidney diseases of unknown origin where molecular genetic testing is lacking. However, while hypomagnesaemia, in the absence of diuretics, appears to rule out ADPKD and ARPKD, normomagnesaemia does not rule out RCAD at least in those aged <3 years.


2020 ◽  
Vol 15 (10) ◽  
pp. 1497-1510 ◽  
Author(s):  
Enrico Cocchi ◽  
Jordan Gabriela Nestor ◽  
Ali G. Gharavi

Expanded accessibility of genetic sequencing technologies, such as chromosomal microarray and massively parallel sequencing approaches, is changing the management of hereditary kidney diseases. Genetic causes account for a substantial proportion of pediatric kidney disease cases, and with increased utilization of diagnostic genetic testing in nephrology, they are now also detected at appreciable frequencies in adult populations. Establishing a molecular diagnosis can have many potential benefits for patient care, such as guiding treatment, familial testing, and providing deeper insights on the molecular pathogenesis of kidney diseases. Today, with wider clinical use of genetic testing as part of the diagnostic evaluation, nephrologists have the challenging task of selecting the most suitable genetic test for each patient, and then applying the results into the appropriate clinical contexts. This review is intended to familiarize nephrologists with the various technical, logistical, and ethical considerations accompanying the increasing utilization of genetic testing in nephrology care.


2019 ◽  
Vol 51 (1) ◽  
pp. 1-10 ◽  
Author(s):  
Xin Li ◽  
Ming Wu ◽  
Limin Chen ◽  
Junyan Lu ◽  
Guo Li ◽  
...  

Background: Inflammation plays an important role in polycystic kidney disease (PKD). Cordyceps sinensis, a prized ­Chinese medicinal herb, exerts anti-tumor, anti-inflammatory and anti-metastatic effects and benefits patients with kidney diseases. The aim of this study was to test the efficacy of FTY720, an immunosuppressant derived from C. sinensis, in a rat cystic kidney disease model, and explore its underlining mechanism. Methods: Male wild type and Cy/+ Han:SPRD rats were treated with FTY720 at 3 and 10 mg/kg/day for 5 weeks and 12 weeks by gavage. Blood and kidney were collected for functional, morphological, RNA, and protein analysis. Results: Inflammation is activated in Cy/+ Han:SPRD rats. Inflammatory cytokines including interleukin 6 and tumor necrosis factor alpha were upregulated and inflammation-related pathways were activated, such as nuclear factor κB and signal transducer and activator of transcription 3 (STAT3) pathways. Furthermore, the bioactive sphingolipid mediator sphingosine-1-phosphate (S1P), a regulator of inflammation, was accumulated in the Cy/+ Han:SPRD rats. FTY720 significantly reduced cyst growth and delayed disease progression by reducing the accumulation of S1P, thereby inhibiting inflammatory responses. Conclusion: FTY720 treatment reduced the expression of inflammatory cytokines and attenuated the activation of NK-κB and STAT3 pathways in Cy/+ Han:SPRD rats. It suggests that FTY720 may serve as a therapeutic agent for clinical autosomal dominant PKD treatment.


2020 ◽  
Vol 35 (Supplement_3) ◽  
Author(s):  
Claudia Izzi ◽  
Elisa Delbarba ◽  
Laura Econimo ◽  
Chiara Dordoni ◽  
Gianfranco Savoldi ◽  
...  

Abstract Background and Aims Discordant affected relative-pairs are seen in ∼10% of families with Autosomal Dominant Polycystic Kidney Disease (ADPKD); &lt;1% of patients exhibit very early onset (VEO) disease. Complex genotypes may result in renal disease variability beyond that predicted by the sole effect of a single PKD mutant allele, leading to the discovery of biallelic or digenic disease. Here we illustrate such complexity in 6 ADPKD pedigrees. Method Among our single-center ADPKD cohort (186 index patients), we selected pedigrees (P) in which marked familial phenotypic variability or severe and early onset disease was investigated by NGS and MLPA analysis of PKD1 and PKD2 genes and NGS analysis of other cystogenes. Segregation analysis by Sanger sequencing of PKD variants was performed in available affected and unaffected family members. Results In P1 and P2, the index cases (IC), presented with very early onset (VEO) disease characterized by prenatal/neonatal enlarged and hyperechogenic kidneys mimicking autosomal recessive polycystic kidney disease (ARPKD). In P1, with neonatal onset, the ADPKD affected father transmitted a PKD1 PT variant p.Gln4231*, whereas the mother, without renal cystic phenotype, transmitted a PKD1 hypomorphic variant p.Asp1332Asn. In P2, the ADPKD-PKD2 mother’s pregnancy was complicated by Potter sequence. Parent’s PKHD1 gene analysis was negative. Two missense NT variants in PKD1/PKD2 genes were detected in the healthy father, respectively p.Gly1944Arg and p.Thr203Ile. Therefore, a complex PKD inheritance was supposed in the fetus. Fetus DNA was not available. In P3 early onset (EO) ADPKD in two monozygous twins was underpinned by a PKD1 NT variant (p.Arg1951Gln) inherited by the ADPKD mild affected father and worsened by a de novo PKD1 truncating variant p.Arg2402*. In P4 and P5 a digenic ADPKD (PKD1 +PKD2 and PKD1 +PKHD1) was diagnosed in severe ADPKD IC. In P4 the two most severely affected siblings carried a PKD2 T variant (p.Ala365fs) and a PKD1 NT variant p-Cys259Tyr. In P5 the IC presented with EO ADPKD, a de novo splicing variant c.2097 + 5_+6insT in PKD1 gene was discovered but the phenotype was probably worsened by the presence of biallelic variant in a second cystogene PKHD1: one paternally inherited: p.Gly1712Arg and one maternally inherited: p.Asp3088Asn . Elderly parents in P6 had mild ADPKD with bilateral few kidney cysts and preserved eGFR, whereas IC showed moderate/severe CKD due to ADPKD biallelic variants. The IC carried a homozygous PKD1 NT variant (p.Arg4154Cys): each mutant allele inherited from the mild ADPKD affected parents. Conclusion Our study illustrates the genetic complexity in an otherwise “simple” Mendelian disorder, providing insights into the genetic basis of severity of ADPKD cases and into ADPKD intrafamilial disease variability. In our pedigree all cases with more severe clinical picture in the family presented at least two PKD variants. In P5 we found for the first time an EO ADPKD due to both PKD1 and PKHD1 variants. PKD1 and PKD2 sequence analysis together with cystic kidney disease gene panel analysis is recommended in those patients with discordant phenotype compared to family members. Molecular study of PKD patients is expected to be a good prognostic tool together with clinical and renal imaging data to better manage disease therapy, follow-up and reproductive issues.


Sign in / Sign up

Export Citation Format

Share Document