1
|
Paliwal P, Thakur S, Sharma S. Bright and enlarged fetal kidneys: One phenotype different genotypes, and counseling dilemmas. JOURNAL OF CLINICAL ULTRASOUND : JCU 2024; 52:1370-1374. [PMID: 39234693 DOI: 10.1002/jcu.23797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/11/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
INTRODUCTION In the present study we describe atypical cases with bright and enlarged fetal kidneys identified on fetal ultrasound with different genetic etiologies. METHODS Exome sequencing was undertaken after prenatal counseling and after the initial diagnosis of enlarged fetal kidneys was made on ultrasound for four cases and the results were then correlated. RESULTS In the present study we identified underlying variants in ACE, ETFA, PKD1, and MKS1 gene where the atypical presentation of fetal kidneys was noted either as a part of spectrum of syndrome or alone. CONCLUSIONS In the era of exome sequencing, targeted gene sequencing is getting replaced and for better. However not all answers are direct, and sometimes the variant categorization is dependent on the acumen and agreement of all those involved in the process. It includes those involved the diagnostic as well those catering to the patients. It is very important to be updated on the relevance of multiple gene in causing similar phenotypes particularly in the prenatal context were coming up with a timely diagnosis is very important for any sort of intervention.
Collapse
Affiliation(s)
| | - Seema Thakur
- Fortis Hospitals, The Genetic clinic, New Delhi, India
| | | |
Collapse
|
2
|
Stark Z, Byrne AB, Sampson MG, Lennon R, Mallett AJ. A guide to gene-disease relationships in nephrology. Nat Rev Nephrol 2024:10.1038/s41581-024-00900-7. [PMID: 39443743 DOI: 10.1038/s41581-024-00900-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2024] [Indexed: 10/25/2024]
Abstract
The use of next-generation sequencing technologies such as exome and genome sequencing in research and clinical care has transformed our understanding of the molecular architecture of genetic kidney diseases. Although the capability to identify and rigorously assess genetic variants and their relationship to disease has advanced considerably in the past decade, the curation of clinically relevant relationships between genes and specific phenotypes has received less attention, despite it underpinning accurate interpretation of genomic tests. Here, we discuss the need to accurately define gene-disease relationships in nephrology and provide a framework for appraising genetic and experimental evidence critically. We describe existing international programmes that provide expert curation of gene-disease relationships and discuss sources of discrepancy as well as efforts at harmonization. Further, we highlight the need for alignment of disease and phenotype terminology to ensure robust and reproducible curation of knowledge. These collective efforts to support evidence-based translation of genomic sequencing into practice across clinical, diagnostic and research settings are crucial for delivering the promise of precision medicine in nephrology, providing more patients with timely diagnoses, accurate prognostic information and access to targeted treatments.
Collapse
Affiliation(s)
- Zornitza Stark
- ClinGen, Boston, MA, USA.
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.
- Australian Genomics, Melbourne, Victoria, Australia.
| | - Alicia B Byrne
- ClinGen, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Matthew G Sampson
- ClinGen, Boston, MA, USA
- Division of Nephrology, Boston Children's Hospital, Boston, MA, USA
- Department of Paediatrics, Harvard Medical School, Boston, MA, USA
| | - Rachel Lennon
- ClinGen, Boston, MA, USA
- Wellcome Centre for Cell-Matrix Research, The University of Manchester, Manchester, UK
- Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester, UK
| | - Andrew J Mallett
- ClinGen, Boston, MA, USA.
- Townsville Hospital and Health Service, Townsville, Queensland, Australia.
- College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia.
- Institute for Molecular Bioscience and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
3
|
Bracciamà V, Vaisitti T, Mioli F, Faini AC, Del Prever GMB, Martins VH, Camilla R, Mattozzi F, Pieretti S, Luca M, Romeo CM, Saglia C, Migliorero M, Arruga F, Carli D, Amoroso A, Lonardi P, Deaglio S, Peruzzi L. Matching clinical and genetic data in pediatric patients at risk of developing cystic kidney disease. Pediatr Nephrol 2024:10.1007/s00467-024-06548-6. [PMID: 39384646 DOI: 10.1007/s00467-024-06548-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/11/2024] [Accepted: 09/27/2024] [Indexed: 10/11/2024]
Abstract
BACKGROUND Cystic kidney disease is a heterogeneous group of hereditary and non-hereditary pathologic conditions, associated with the development of renal cysts. These conditions may be present both in children and adults. Cysts can even be observed already during the prenatal age, and pediatric patients with cysts need to be clinically monitored. An early clinical and genetic diagnosis is therefore mandatory for optimal patient management. The aim of this study was to perform genetic analyses in patients with echographic evidence of kidney cysts to provide an early molecular diagnosis. METHODS A cohort of 70 pediatric patients was enrolled and clinically studied at the time of first recruitment and at follow-up. Genetic testing by clinical exome sequencing was performed and a panel of genes responsible for "cystic kidneys" was analyzed to identify causative variants. Sanger validation and segregation studies were exploited for the final classification of the variants and accurate genetic counseling. RESULTS Data showed that 53/70 of pediatric patients referred with a clinical suspicion of cystic kidney disease presented a causative genetic variant. In a significant proportion of the cohort (24/70), evidence of hyper-echogenic/cystic kidneys was already present in the prenatal period, even in the absence of a positive family history. CONCLUSIONS This study suggests that cystic kidney disease may develop since the very early stages of life and that screening programs based on ultrasound scans and genetic testing play a critical role in diagnosis, allowing for better clinical management and tailored genetic counseling to the family.
Collapse
Affiliation(s)
- Valeria Bracciamà
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Tiziana Vaisitti
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy.
| | - Fiorenza Mioli
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Angelo Corso Faini
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Giulia Margherita Brach Del Prever
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vitor Hugo Martins
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Roberta Camilla
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Francesca Mattozzi
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Silvia Pieretti
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Maria Luca
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Carmelo Maria Romeo
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Claudia Saglia
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Martina Migliorero
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Francesca Arruga
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Diana Carli
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Antonio Amoroso
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Pietro Lonardi
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| | - Silvia Deaglio
- Immunogenetics and Transplant Biology, AOU Città della Salute e della Scienza, ERKNet Center & Department of Medical Sciences, University of Turin, Turin, Italy
| | - Licia Peruzzi
- Nephrology Dialysis and Transplantation, ERKNet Center, Regina Margherita Children's Hospital, Turin, Italy
| |
Collapse
|
4
|
Tran U, Streets AJ, Smith D, Decker E, Kirschfink A, Izem L, Hassey JM, Rutland B, Valluru MK, Bräsen JH, Ott E, Epting D, Eisenberger T, Ong AC, Bergmann C, Wessely O. BICC1 Interacts with PKD1 and PKD2 to Drive Cystogenesis in ADPKD. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.608867. [PMID: 39253489 PMCID: PMC11383298 DOI: 10.1101/2024.08.27.608867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is primarily of adult-onset and caused by pathogenic variants in PKD1 or PKD2 . Yet, disease expression is highly variable and includes very early-onset PKD presentations in utero or infancy. In animal models, the RNA-binding molecule Bicc1 has been shown to play a crucial role in the pathogenesis of PKD. Methods To study the interaction between BICC1, PKD1 and PKD2 we combined biochemical approaches, knockout studies in mice and Xenopus, genetic engineered human kidney cells as well as genetic association studies in a large ADPKD cohort. Results We first demonstrated that BICC1 physically binds to the proteins Polycystin-1 and -2 encoded by PKD1 and PKD2 via distinct protein domains. Furthermore, PKD was aggravated in loss-of-function studies in Xenopus and mouse models resulting in more severe disease when Bicc1 was depleted in conjunction with Pkd1 or Pkd2 . Finally, in a large human patient cohort, we identified a sibling pair with a homozygous BICC1 variant and patients with very early onset PKD (VEO-PKD) that exhibited compound heterozygosity of BICC1 in conjunction with PKD1 and PKD2 variants. Genome editing demonstrated that these BICC1 variants were hypomorphic in nature and impacted disease-relevant signaling pathways. Conclusions These findings support the hypothesis that BICC1 cooperates functionally with PKD1 and PKD2 , and that BICC1 variants may aggravate disease severity highlighting RNA metabolism as an important new concept for disease modification in ADPKD.
Collapse
|
5
|
Vivante A. Genetics of Chronic Kidney Disease. N Engl J Med 2024; 391:627-639. [PMID: 39141855 DOI: 10.1056/nejmra2308577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Affiliation(s)
- Asaf Vivante
- From the Department of Pediatrics and the Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, and the Nephro-Genetics Clinic and Genetic Kidney Disease Research Laboratory, Sheba Medical Center, Tel Hashomer, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - all in Israel
| |
Collapse
|
6
|
Chen EWC, Chong J, Valluru MK, Durkie M, Simms RJ, Harris PC, Ong ACM. Combining genotype with height-adjusted kidney length predicts rapid progression of ADPKD. Nephrol Dial Transplant 2024; 39:956-966. [PMID: 38224954 DOI: 10.1093/ndt/gfad270] [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: 06/29/2023] [Indexed: 01/17/2024] Open
Abstract
INTRODUCTION Our main objective was to identify baseline prognostic factors predictive of rapid disease progression in a large unselected clinical autosomal dominant polycystic kidney disease (ADPKD) cohort. METHODS A cross-sectional analysis was performed in 618 consecutive ADPKD patients assessed and followed-up for over a decade. A total of 123 patients (19.9%) had reached kidney failure by the study date. Data were available for the following: baseline eGFR (n = 501), genotype (n = 549), baseline ultrasound mean kidney length (MKL, n = 424) and height-adjusted baseline MKL (HtMKL, n = 377). Rapid disease progression was defined as an annualized eGFR decline (∆eGFR) of >2.5 mL/min/year by linear regression over 5 years (n = 158). Patients were further divided into slow, rapid and very rapid ∆eGFR classes for analysis. Genotyped patients were classified into several categories: PKD1 (T, truncating; or NT, non-truncating), PKD2, other genes (non-PKD1 or -PKD2), no mutation detected or variants of uncertain significance. RESULTS A PKD1-T genotype had the strongest influence on the probability of reduced baseline kidney function by age. A multivariate logistic regression model identified PKD1-T genotype and HtMKL (>9.5 cm/m) as independent predictors for rapid disease progression. The combination of both factors increased the positive predictive value for rapid disease progression over age 40 years and of reaching kidney failure by age 60 years to 100%. Exploratory analysis in a subgroup with available total kidney volumes showed higher positive predictive value (100% vs 80%) and negative predictive value (42% vs 33%) in predicting rapid disease progression compared with the Mayo Imaging Classification (1C-E). CONCLUSION Real-world longitudinal data confirm the importance of genotype and kidney length as independent variables determining ∆eGFR. Individuals with the highest risk of rapid disease progression can be positively selected for treatment based on this combination.
Collapse
Affiliation(s)
- Eugene W C Chen
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, UK
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Herries Road, Sheffield, UK
| | - Jiehan Chong
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, UK
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Herries Road, Sheffield, UK
| | - Manoj K Valluru
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, UK
| | - Miranda Durkie
- Sheffield Diagnostics Genetic Service, North East and Yorkshire Genomic Laboratory Hub, Sheffield Children's NHS Foundation Trust, Sheffield, UK
| | - Roslyn J Simms
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, UK
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Herries Road, Sheffield, UK
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic and Foundation, Rochester, MN, USA
| | - Albert C M Ong
- Academic Nephrology Unit, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road, Sheffield, UK
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Herries Road, Sheffield, UK
| |
Collapse
|
7
|
Thuma TBT, Procopio RA, Jimenez HJ, Gunton KB, Pulido JS. Hypomorphic variants in inherited retinal and ocular diseases: A review of the literature with clinical cases. Surv Ophthalmol 2024; 69:337-348. [PMID: 38036193 DOI: 10.1016/j.survophthal.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 11/17/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
Hypomorphic variants decrease, but do not eliminate, gene function via a reduction in the amount of mRNA or protein product produced by a gene or by production of a gene product with reduced function. Many hypomorphic variants have been implicated in inherited retinal diseases (IRDs) and other genetic ocular conditions; however, there is heterogeneity in the use of the term "hypomorphic" in the scientific literature. We searched for all hypomorphic variants reported to cause IRDs and ocular disorders. We also discuss the presence of hypomorphic variants in the patient population of our ocular genetics department over the past decade. We propose that standardized criteria should be adopted for use of the term "hypomorphic" to describe gene variants to improve genetic counseling and patient care outcomes.
Collapse
Affiliation(s)
- Tobin B T Thuma
- Department of Pediatric Ophthalmology and Strabismus, Wills Eye Hospital, Philadelphia, PA, USA
| | | | - Hiram J Jimenez
- Vickie and Jack Farber Vision Research Center, Wills Eye Hospital, Philadelphia, PA, USA
| | - Kammi B Gunton
- Department of Pediatric Ophthalmology and Strabismus, Wills Eye Hospital, Philadelphia, PA, USA
| | - Jose S Pulido
- Vickie and Jack Farber Vision Research Center, Wills Eye Hospital, Philadelphia, PA, USA; Retina Service, Wills Eye Hospital, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Ali H, Alahmad B, Senum SR, Warsame S, Bahbahani Y, Abu-Farha M, Abubaker J, Alqaddoumi M, Al-Mulla F, Harris PC. PKD1 Truncating Mutations Accelerate eGFR Decline in Autosomal Dominant Polycystic Kidney Disease Patients. Am J Nephrol 2024; 55:380-388. [PMID: 38194940 PMCID: PMC11151966 DOI: 10.1159/000536165] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 01/04/2024] [Indexed: 01/11/2024]
Abstract
INTRODUCTION Autosomal dominant polycystic kidney disease (ADPKD) is a monogenic disease characterized by the accumulation of fluid-filled cysts in the kidneys, leading to renal volume enlargement and progressive kidney function impairment. Disease severity, though, may vary due to allelic and genetic heterogeneity. This study aimed to determine genotype-phenotype correlations between PKD1 truncating and non-truncating mutations and kidney function decline in ADPKD patients. METHODS We established a single-center retrospective cohort study in Kuwait where we followed every patient with a confirmed PKD1-ADPKD diagnosis clinically and genetically. Renal function tests were performed annually. We fitted generalized additive mixed effects models with random intercepts for each individual to analyze repeated measures of kidney function across mutation type. We then calculated survival time to kidney failure in a cox proportional hazards model. Models were adjusted for sex, age at visit, and birth year. RESULTS The study included 22 truncating and 20 non-truncating (42 total) patients followed for an average of 6.6 years (range: 1-12 years). Those with PKD1 truncating mutations had a more rapid rate of eGFR decline (-4.7 mL/min/1.73 m2 per year; 95% CI: -5.0, -4.4) compared to patients with PKD1 non-truncating mutations (-3.5 mL/min/1.73 m2 per year; 95% CI: -4.0, -3.1) (p for interaction <0.001). Kaplan-Meier survival analysis of time to kidney failure showed that patients with PKD1 truncating mutations had a shorter renal survival time (median 51 years) compared to those with non-truncating mutations (median 56 years) (P for log-rank = 0.008). CONCLUSION In longitudinal and survival analyses, patients with PKD1 truncating mutations showed a faster decline in kidney function compared to patients PKD1 non-truncating mutations. Early identification of patients with PKD1 truncating mutations can, at best, inform early clinical interventions or, at least, help suggest aggressive monitoring.
Collapse
Affiliation(s)
- Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Barrak Alahmad
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Sarah R. Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Department of Artificial Intelligence and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Samia Warsame
- Medical Division, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Yousif Bahbahani
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
- Medical Division, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Malak Alqaddoumi
- Department of Pathology, Faculty of Medicine, Health Sciences Center (HSC), Kuwait University, Jabriya, Kuwait
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
9
|
Yang H, Sieben CJ, Schauer RS, Harris PC. Genetic Spectrum of Polycystic Kidney and Liver Diseases and the Resulting Phenotypes. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:397-406. [PMID: 38097330 PMCID: PMC10746289 DOI: 10.1053/j.akdh.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/21/2023] [Accepted: 04/27/2023] [Indexed: 12/18/2023]
Abstract
Polycystic kidney diseases are a group of monogenically inherited disorders characterized by cyst development in the kidney with defects in primary cilia function central to pathogenesis. Autosomal dominant polycystic kidney disease (ADPKD) has progressive cystogenesis and accounts for 5-10% of kidney failure (KF) patients. There are two major ADPKD genes, PKD1 and PKD2, and seven minor loci. PKD1 accounts for ∼80% of patients and is associated with the most severe disease (KF is typically at 55-65 years); PKD2 accounts for ∼15% of families, with KF typically in the mid-70s. The minor genes are generally associated with milder kidney disease, but for DNAJB11 and ALG5, the age at KF is similar to PKD2. PKD1 and PKD2 have a high level of allelic heterogeneity, with no single pathogenic variant accounting for >2% of patients. Additional genetic complexity includes biallelic disease, sometimes causing very early-onset ADPKD, and mosaicism. Autosomal dominant polycystic liver disease is characterized by severe PLD but limited PKD. The two major genes are PRKCSH and SEC63, while GANAB, ALG8, and PKHD1 can present as ADPKD or autosomal dominant polycystic liver disease. Autosomal recessive polycystic kidney disease typically has an infantile onset, with PKHD1 being the major locus and DZIP1L and CYS1 being minor genes. In addition, there are a range of mainly recessive syndromic ciliopathies with PKD as part of the phenotype. Because of the phenotypic and genic overlap between the diseases, employing a next-generation sequencing panel containing all known PKD and ciliopathy genes is recommended for clinical testing.
Collapse
Affiliation(s)
- Hana Yang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester MN
| | - Cynthia J Sieben
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester MN
| | - Rachel S Schauer
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester MN
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester MN.
| |
Collapse
|
10
|
Burgmaier K, Broekaert IJ, Liebau MC. Autosomal Recessive Polycystic Kidney Disease: Diagnosis, Prognosis, and Management. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:468-476. [PMID: 38097335 DOI: 10.1053/j.akdh.2023.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 12/18/2023]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD) is the rare and usually early-onset form of polycystic kidney disease with a typical clinical presentation of enlarged cystic kidneys and liver involvement with congenital hepatic fibrosis or Caroli syndrome. ARPKD remains a clinical challenge in pediatrics, frequently requiring continuous and long-term multidisciplinary treatment. In this review, we aim to give an overview over clinical aspects of ARPKD and recent developments in our understanding of disease progression, risk patterns, and treatment of ARPKD.
Collapse
Affiliation(s)
- Kathrin Burgmaier
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany; Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Ilse J Broekaert
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Max C Liebau
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany; Center for Family Health, Center for Rare Diseases and Center for Molecular Medicine Cologne, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
11
|
Lin Y, Chen X, Liang C, Li D, Liu L, Li X. Novel compound heterozygous variant of GJA8 gene in two siblings with congenital cataract mimics an autosomal recessive trait. Eur J Ophthalmol 2023; 33:NP1-NP4. [PMID: 36262071 DOI: 10.1177/11206721221132874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND GJA8 gene is known to cause autosomal dominant congenital cataract. Here we report a novel compound heterozygous variant of GJA8 gene in two siblings that mimics an autosomal recessive trait. PATIENTS AND METHODS Two siblings from a non-consanguineous Chinese family suffered from isolated congenital cataract. Whole exome sequencing was performed to identify disease-causing variants followed by a confirmatory Sanger sequencing. RESULTS Whole exome sequencing revealed a novel compound heterozygous variant of GJA8 gene, c.855delG(p.Met286Trpfs*71)/c.1125delC(p.Gly376Glufs*33), in the proband. Sanger sequencing confirmed that the proband and his sister harboured this compound heterozygous variant, while the parents were heterozygous carriers, suggesting an autosomal recessive inheritance pattern. Both parents showed mildly impaired vision, but only the father had mild nuclear opacities, suggesting an autosomal dominant trait with reduced penetrance in one of the pathogenic alleles. CONCLUSIONS Our report shows compound heterozygous variant of GJA8 gene may mimic autosomal recessive inheritance pattern, and reminds clinicians to perform needful examination. The two novel pathogenic GJA8 variants expand the mutational spectrum of congenital cataract. This study also provides accurate genetic diagnosis for the family.
Collapse
Affiliation(s)
- Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Xiaodan Chen
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Cuili Liang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Duan Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| | - Xiuzhen Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou 510623, China
| |
Collapse
|
12
|
Graziani L, Zampatti S, Carriero ML, Minotti C, Peconi C, Bengala M, Giardina E, Novelli G. Co-Inheritance of Pathogenic Variants in PKD1 and PKD2 Genes Determined by Parental Segregation and De Novo Origin: A Case Report. Genes (Basel) 2023; 14:1589. [PMID: 37628640 PMCID: PMC10454652 DOI: 10.3390/genes14081589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/01/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary renal disease, and it is typically caused by PKD1 and PKD2 heterozygous variants. Nonetheless, the extensive phenotypic variability observed among affected individuals, even within the same family, suggests a more complex pattern of inheritance. We describe an ADPKD family in which the proband presented with an earlier and more severe renal phenotype (clinical diagnosis at the age of 14 and end-stage renal disease aged 24), compared to the other affected family members. Next-generation sequencing (NGS)-based analysis of polycystic kidney disease (PKD)-associated genes in the proband revealed the presence of a pathogenic PKD2 variant and a likely pathogenic variant in PKD1, according to the American College of Medical Genetics and Genomics (ACMG) criteria. The PKD2 nonsense p.Arg872Ter variant was segregated from the proband's father, with a mild phenotype. A similar mild disease presentation was found in the proband's aunts and uncle (the father's siblings). The frameshift p.Asp3832ProfsTer128 novel variant within PKD1 carried by the proband in addition to the pathogenic PKD2 variant was not found in either parent. This report highlights that the co-inheritance of two or more PKD genes or alleles may explain the extensive phenotypic variability among affected family members, thus emphasizing the importance of NGS-based techniques in the definition of the prognostic course.
Collapse
Affiliation(s)
- Ludovico Graziani
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.L.C.); (C.M.); (E.G.); (G.N.)
| | - Stefania Zampatti
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.)
| | - Miriam Lucia Carriero
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.L.C.); (C.M.); (E.G.); (G.N.)
| | - Chiara Minotti
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.L.C.); (C.M.); (E.G.); (G.N.)
| | - Cristina Peconi
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.)
| | - Mario Bengala
- Medical Genetics Unit, Tor Vergata University Hospital, 00133 Rome, Italy;
| | - Emiliano Giardina
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.L.C.); (C.M.); (E.G.); (G.N.)
- Genomic Medicine Laboratory UILDM, IRCCS Fondazione Santa Lucia, 00179 Rome, Italy; (S.Z.); (C.P.)
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, 00133 Rome, Italy; (M.L.C.); (C.M.); (E.G.); (G.N.)
- Medical Genetics Unit, Tor Vergata University Hospital, 00133 Rome, Italy;
| |
Collapse
|
13
|
Halawi AA, Burgmaier K, Buescher AK, Dursun I, Erger F, Galiano M, Gessner M, Gökce I, Mekahli D, Mir S, Obrycki L, Shroff R, Stabouli S, Szczepanska M, Teixeira A, Weber LT, Wenzel A, Wühl E, Zachwieja K, Dötsch J, Schaefer F, Liebau MC. Clinical Characteristics and Courses of Patients With Autosomal Recessive Polycystic Kidney Disease-Mimicking Phenocopies. Kidney Int Rep 2023; 8:1449-1454. [PMID: 37441483 PMCID: PMC10334384 DOI: 10.1016/j.ekir.2023.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 03/06/2023] [Accepted: 04/04/2023] [Indexed: 07/15/2023] Open
Affiliation(s)
- Abdul A. Halawi
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Kathrin Burgmaier
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Anja K. Buescher
- Department of Pediatrics II, University Hospital Essen, Essen, Germany
| | - Ismail Dursun
- Department of Pediatric Nephrology, Erciyes University, Faculty of Medicine, Kayseri, Turkey
| | - Florian Erger
- Institute of Human Genetics, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Rare Diseases, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Matthias Galiano
- Department of Pediatrics and Adolescent Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Michaela Gessner
- Department of General Pediatrics and Hematology/ Oncology, Children’s University Hospital Tuebingen, Germany
| | - Ibrahim Gökce
- Research and Training Hospital, Division of Pediatric Nephrology, Marmara University, Istanbul, Turkey
| | - Djalila Mekahli
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, PKD Research Group, KU Leuven, Leuven, Belgium
| | - Sevgi Mir
- Department of Pediatric Nephrology, Ege University Medical Faculty, Izmir, Turkey
| | - Lukasz Obrycki
- Department of Nephrology, Kidney Transplantation and Hypertension, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Rukshana Shroff
- UCL Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Stella Stabouli
- First Department of Pediatrics, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Maria Szczepanska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, SUM in Katowice, Katowice, Poland
| | - Ana Teixeira
- Pediatric Nephrology, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Lutz T. Weber
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Andrea Wenzel
- Institute of Human Genetics, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Rare Diseases, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Elke Wühl
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Katarzyna Zachwieja
- Department of Pediatric Nephrology and Hypertension Jagiellonian University Medical College, Krakow, Poland
| | - Jörg Dötsch
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Rare Diseases, University Hospital Cologne and University of Cologne, Cologne, Germany
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Max C. Liebau
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, University Hospital Cologne and University of Cologne, Cologne, Germany
- Center for Rare Diseases, University Hospital Cologne and University of Cologne, Cologne, Germany
| |
Collapse
|
14
|
Ambrosini E, Montanari F, Cristalli CP, Capelli I, La Scola C, Pasini A, Graziano C. Modifiers of Autosomal Dominant Polycystic Kidney Disease Severity: The Role of PKD1 Hypomorphic Alleles. Genes (Basel) 2023; 14:1230. [PMID: 37372410 DOI: 10.3390/genes14061230] [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: 04/29/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic cause of kidney failure in adult life. Rarely, ADPKD can be diagnosed in utero or in infancy, and the genetic mechanism underlying such severe presentation has been shown to be related to reduced gene dosage. Biallelic PKD1 variants are often identified in early onset ADPKD, with one main pathogenic variant and a modifier hypomorphic variant showing an in trans configuration. We describe two unrelated individuals with early onset cystic kidney disease and unaffected parents, where a combination of next-generation sequencing of cystic genes including PKHD1, HNF1B and PKD1 allowed the identification of biallelic PKD1 variants. Furthermore, we review the medical literature in order to report likely PKD1 hypomorphic variants reported to date and estimate a minimal allele frequency of 1/130 for this category of variants taken as a group. This figure could help to orient genetic counseling, although the interpretation and the real clinical impact of rare PKD1 missense variants, especially if previously unreported, remain challenging.
Collapse
Affiliation(s)
| | - Francesca Montanari
- Medical Genetics Unit, IRCCS Sant'Orsola University Hospital of Bologna, 40138 Bologna, Italy
| | - Carlotta Pia Cristalli
- Medical Genetics Unit, IRCCS Sant'Orsola University Hospital of Bologna, 40138 Bologna, Italy
| | - Irene Capelli
- Nephrology Unit, IRCCS Sant'Orsola University Hospital of Bologna, 40138 Bologna, Italy
| | - Claudio La Scola
- Paediatric Nephrology Program, Paediatrics Unit, IRCCS Sant'Orsola University Hospital of Bologna, 40138 Bologna, Italy
| | - Andrea Pasini
- Paediatric Nephrology Program, Paediatrics Unit, IRCCS Sant'Orsola University Hospital of Bologna, 40138 Bologna, Italy
| | | |
Collapse
|
15
|
Claus LR, Snoek R, Knoers NVAM, van Eerde AM. Review of genetic testing in kidney disease patients: Diagnostic yield of single nucleotide variants and copy number variations evaluated across and within kidney phenotype groups. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:358-376. [PMID: 36161467 PMCID: PMC9828643 DOI: 10.1002/ajmg.c.31995] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/02/2022] [Accepted: 08/18/2022] [Indexed: 01/29/2023]
Abstract
Genetic kidney disease comprises a diverse group of disorders. These can roughly be divided in the phenotype groups congenital anomalies of the kidney and urinary tract, ciliopathies, glomerulopathies, stone disorders, tubulointerstitial kidney disease, and tubulopathies. Many etiologies can lead to chronic kidney disease that can progress to end-stage kidney disease. Despite each individual disease being rare, together these genetic disorders account for a large proportion of kidney disease cases. With the introduction of massively parallel sequencing, genetic testing has become more accessible, but a comprehensive analysis of the diagnostic yield is lacking. This review gives an overview of the diagnostic yield of genetic testing across and within the full range of kidney disease phenotypes through a systematic literature search that resulted in 115 included articles. Patient, test, and cohort characteristics that can influence the diagnostic yield are highlighted. Detection of copy number variations and their contribution to the diagnostic yield is described for all phenotype groups. Also, the impact of a genetic diagnosis for a patient and family members, which can be diagnostic, therapeutic, and prognostic, is shown through the included articles. This review will allow clinicians to estimate an a priori probability of finding a genetic cause for the kidney disease in their patients.
Collapse
Affiliation(s)
- Laura R. Claus
- Department of GeneticsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Rozemarijn Snoek
- Department of GeneticsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Nine V. A. M. Knoers
- Department of GeneticsUniversity Medical Center GroningenGroningenThe Netherlands
| | | |
Collapse
|
16
|
Masson E, Zou WB, Génin E, Cooper DN, Le Gac G, Fichou Y, Pu N, Rebours V, Férec C, Liao Z, Chen JM. Expanding ACMG variant classification guidelines into a general framework. Hum Genomics 2022; 16:31. [PMID: 35974416 PMCID: PMC9380380 DOI: 10.1186/s40246-022-00407-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 08/10/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The American College of Medical Genetics and Genomics (ACMG)-recommended five variant classification categories (pathogenic, likely pathogenic, uncertain significance, likely benign, and benign) have been widely used in medical genetics. However, these guidelines are fundamentally constrained in practice owing to their focus upon Mendelian disease genes and their dichotomous classification of variants as being either causal or not. Herein, we attempt to expand the ACMG guidelines into a general variant classification framework that takes into account not only the continuum of clinical phenotypes, but also the continuum of the variants' genetic effects, and the different pathological roles of the implicated genes. MAIN BODY As a disease model, we employed chronic pancreatitis (CP), which manifests clinically as a spectrum from monogenic to multifactorial. Bearing in mind that any general conceptual proposal should be based upon sound data, we focused our analysis on the four most extensively studied CP genes, PRSS1, CFTR, SPINK1 and CTRC. Based upon several cross-gene and cross-variant comparisons, we first assigned the different genes to two distinct categories in terms of disease causation: CP-causing (PRSS1 and SPINK1) and CP-predisposing (CFTR and CTRC). We then employed two new classificatory categories, "predisposing" and "likely predisposing", to replace ACMG's "pathogenic" and "likely pathogenic" categories in the context of CP-predisposing genes, thereby classifying all pathologically relevant variants in these genes as "predisposing". In the case of CP-causing genes, the two new classificatory categories served to extend the five ACMG categories whilst two thresholds (allele frequency and functional) were introduced to discriminate "pathogenic" from "predisposing" variants. CONCLUSION Employing CP as a disease model, we expand ACMG guidelines into a five-category classification system (predisposing, likely predisposing, uncertain significance, likely benign, and benign) and a seven-category classification system (pathogenic, likely pathogenic, predisposing, likely predisposing, uncertain significance, likely benign, and benign) in the context of disease-predisposing and disease-causing genes, respectively. Taken together, the two systems constitute a general variant classification framework that, in principle, should span the entire spectrum of variants in any disease-related gene. The maximal compliance of our five-category and seven-category classification systems with the ACMG guidelines ought to facilitate their practical application.
Collapse
Affiliation(s)
- Emmanuelle Masson
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France.,Service de Génétique Médicale et de Biologie de la Reproduction, CHRU Brest, F-29200, Brest, France
| | - Wen-Bin Zou
- Department of Gastroenterology, Changhai Hospital, The Secondary Military Medical University, Shanghai, China.,Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Emmanuelle Génin
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, UK
| | - Gerald Le Gac
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France.,Service de Génétique Médicale et de Biologie de la Reproduction, CHRU Brest, F-29200, Brest, France
| | - Yann Fichou
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France
| | - Na Pu
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France.,Department of Critical Care Medicine, Research Institute of General Surgery, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Vinciane Rebours
- Department of Gastroenterology and Pancreatology, Beaujon Hospital, Assistance Publique-Hôpitaux de Paris, Clichy, Université de Paris, Paris, France
| | - Claude Férec
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France
| | - Zhuan Liao
- Department of Gastroenterology, Changhai Hospital, The Secondary Military Medical University, Shanghai, China.,Shanghai Institute of Pancreatic Diseases, Shanghai, China
| | - Jian-Min Chen
- Univ Brest, Inserm, EFS, UMR 1078, GGB, 22 Avenue Camille Desmoulins, F-29200, Brest, France.
| |
Collapse
|
17
|
Wang JY, Wang J, Lu XG, Song W, Luo S, Zou DF, Hua LD, Peng Q, Tian Y, Gao LD, Liao WP, He N. Recessive PKD1 Mutations Are Associated With Febrile Seizures and Epilepsy With Antecedent Febrile Seizures and the Genotype-Phenotype Correlation. Front Mol Neurosci 2022; 15:861159. [PMID: 35620448 PMCID: PMC9128595 DOI: 10.3389/fnmol.2022.861159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThe PKD1 encodes polycystin-1, a large transmembrane protein that plays important roles in cell proliferation, apoptosis, and cation transport. Previous studies have identified PKD1 mutations in autosomal dominant polycystic kidney disease (ADPKD). However, the expression of PKD1 in the brain is much higher than that in the kidney. This study aimed to explore the association between PKD1 and epilepsy.MethodsTrios-based whole-exome sequencing was performed in a cohort of 314 patients with febrile seizures or epilepsy with antecedent febrile seizures. The damaging effects of variants was predicted by protein modeling and multiple in silico tools. The genotype-phenotype association of PKD1 mutations was systematically reviewed and analyzed.ResultsEight pairs of compound heterozygous missense variants in PKD1 were identified in eight unrelated patients. All patients suffered from febrile seizures or epilepsy with antecedent febrile seizures with favorable prognosis. All of the 16 heterozygous variants presented no or low allele frequencies in the gnomAD database, and presented statistically higher frequency in the case-cohort than that in controls. These missense variants were predicted to be damaging and/or affect hydrogen bonding or free energy stability of amino acids. Five patients showed generalized tonic-clonic seizures (GTCS), who all had one of the paired missense mutations located in the PKD repeat domain, suggesting that mutations in the PKD domains were possibly associated with GTCS. Further analysis demonstrated that monoallelic mutations with haploinsufficiency of PKD1 potentially caused kidney disease, compound heterozygotes with superimposed effects of two missense mutations were associated with epilepsy, whereas the homozygotes with complete loss of PKD1 would be embryonically lethal.ConclusionPKD1 gene was potentially a novel causative gene of epilepsy. The genotype-phenotype relationship of PKD1 mutations suggested a quantitative correlation between genetic impairment and phenotypic variation, which will facilitate the genetic diagnosis and management in patients with PKD1 mutations.
Collapse
Affiliation(s)
- Jing-Yang Wang
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Jie Wang
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Xin-Guo Lu
- Epilepsy Center, Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Wang Song
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Sheng Luo
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Dong-Fang Zou
- Epilepsy Center, Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Li-Dong Hua
- Translational Medicine Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Qian Peng
- Department of Pediatrics, Dongguan City Maternal and Child Health Hospital, Southern Medical University, Dongguan, China
| | - Yang Tian
- Department of Neurology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liang-Di Gao
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wei-Ping Liao
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Na He
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
- *Correspondence: Na He,
| |
Collapse
|
18
|
Peces R, Peces C, Mena R, Cuesta E, García-Santiago FA, Ossorio M, Afonso S, Lapunzina P, Nevado J. Rapidly Progressing to ESRD in an Individual with Coexisting ADPKD and Masked Klinefelter and Gitelman Syndromes. Genes (Basel) 2022; 13:genes13030394. [PMID: 35327948 PMCID: PMC8954516 DOI: 10.3390/genes13030394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/16/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenetic hereditary renal disease, promoting end-stage renal disease (ESRD). Klinefelter syndrome (KS) is a consequence of an extra copy of the X chromosome in males. Main symptoms in KS include hypogonadism, tall stature, azoospermia, and a risk of cardiovascular diseases, among others. Gitelman syndrome (GS) is an autosomal recessive disorder caused by SLC12A3 variants, and is associated with hypokalemia, hypomagnesemia, hypocalciuria, normal or low blood pressure, and salt loss. The three disorders have distinct and well-delineated clinical, biochemical, and genetic findings. We here report a male patient with ADPKD who developed early chronic renal failure leading to ESRD, presenting with an intracranial aneurysm and infertility. NGS identified two de novo PKD1 variants, one known (likely pathogenic), and a previously unreported variant of uncertain significance, together with two SLC12A3 pathogenic variants. In addition, cytogenetic analysis showed a 47, XXY karyotype. We investigated the putative impact of this rare association by analyzing possible clinical, biochemical, and/or genetic interactions and by comparing the evolution of renal size and function in the proband with three age-matched ADPKD (by variants in PKD1) cohorts. We hypothesize that the coexistence of these three genetic disorders may act as modifiers with possible synergistic actions that could lead, in our patient, to a rapid ADPKD progression.
Collapse
Affiliation(s)
- Ramón Peces
- Servicio de Nefrología, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.P.); (M.O.); (S.A.)
| | - Carlos Peces
- Area de Tecnología de la Información, SESCAM, 45071 Toledo, Spain;
| | - Rocío Mena
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.M.); (F.A.G.-S.); (P.L.)
| | - Emilio Cuesta
- Servicio de Radiología, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain;
| | - Fe Amalia García-Santiago
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.M.); (F.A.G.-S.); (P.L.)
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28046 Madrid, Spain
- ITHACA, European Reference Network, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain
| | - Marta Ossorio
- Servicio de Nefrología, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.P.); (M.O.); (S.A.)
| | - Sara Afonso
- Servicio de Nefrología, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.P.); (M.O.); (S.A.)
| | - Pablo Lapunzina
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.M.); (F.A.G.-S.); (P.L.)
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28046 Madrid, Spain
- ITHACA, European Reference Network, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain
| | - Julián Nevado
- Instituto de Genética Médica y Molecular (INGEMM), Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain; (R.M.); (F.A.G.-S.); (P.L.)
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28046 Madrid, Spain
- ITHACA, European Reference Network, Hospital Universitario La Paz, IdiPAZ, Universidad Autónoma, 28046 Madrid, Spain
- Correspondence: ; Tel.: +34-917-277-151; Fax: +34-917-277-382
| |
Collapse
|
19
|
Senum SR, Li Y(SM, Benson KA, Joli G, Olinger E, Lavu S, Madsen CD, Gregory AV, Neatu R, Kline TL, Audrézet MP, Outeda P, Nau CB, Meijer E, Ali H, Steinman TI, Mrug M, Phelan PJ, Watnick TJ, Peters DJ, Ong AC, Conlon PJ, Perrone RD, Cornec-Le Gall E, Hogan MC, Torres VE, Sayer JA, Harris PC, Harris PC. Monoallelic IFT140 pathogenic variants are an important cause of the autosomal dominant polycystic kidney-spectrum phenotype. Am J Hum Genet 2022; 109:136-156. [PMID: 34890546 DOI: 10.1016/j.ajhg.2021.11.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 11/15/2021] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), characterized by progressive cyst formation/expansion, results in enlarged kidneys and often end stage kidney disease. ADPKD is genetically heterogeneous; PKD1 and PKD2 are the common loci (∼78% and ∼15% of families) and GANAB, DNAJB11, and ALG9 are minor genes. PKD is a ciliary-associated disease, a ciliopathy, and many syndromic ciliopathies have a PKD phenotype. In a multi-cohort/-site collaboration, we screened ADPKD-diagnosed families that were naive to genetic testing (n = 834) or for whom no PKD1 and PKD2 pathogenic variants had been identified (n = 381) with a PKD targeted next-generation sequencing panel (tNGS; n = 1,186) or whole-exome sequencing (WES; n = 29). We identified monoallelic IFT140 loss-of-function (LoF) variants in 12 multiplex families and 26 singletons (1.9% of naive families). IFT140 is a core component of the intraflagellar transport-complex A, responsible for retrograde ciliary trafficking and ciliary entry of membrane proteins; bi-allelic IFT140 variants cause the syndromic ciliopathy, short-rib thoracic dysplasia (SRTD9). The distinctive monoallelic phenotype is mild PKD with large cysts, limited kidney insufficiency, and few liver cysts. Analyses of the cystic kidney disease probands of Genomics England 100K showed that 2.1% had IFT140 LoF variants. Analysis of the UK Biobank cystic kidney disease group showed probands with IFT140 LoF variants as the third most common group, after PKD1 and PKD2. The proximity of IFT140 to PKD1 (∼0.5 Mb) in 16p13.3 can cause diagnostic confusion, and PKD1 variants could modify the IFT140 phenotype. Importantly, our studies link a ciliary structural protein to the ADPKD spectrum.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
20
|
Lessons From the Clinic: ADPKD Genetic Test Unraveling Severe Phenotype, Intrafamilial Variability, and New, Rare Causing Genotype. Kidney Int Rep 2022; 7:895-898. [PMID: 35497784 PMCID: PMC9039465 DOI: 10.1016/j.ekir.2021.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/24/2022] Open
|
21
|
The wind of change in the management of autosomal dominant polycystic kidney disease in childhood. Pediatr Nephrol 2022; 37:473-487. [PMID: 33677691 PMCID: PMC8921141 DOI: 10.1007/s00467-021-04974-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/28/2020] [Accepted: 01/27/2021] [Indexed: 12/27/2022]
Abstract
Significant progress has been made in understanding the genetic basis of autosomal dominant polycystic kidney disease (ADPKD), quantifying disease manifestations in children, exploring very-early onset ADPKD as well as pharmacological delay of disease progression in adults. At least 20% of children with ADPKD have relevant, yet mainly asymptomatic disease manifestations such as hypertension or proteinuria (in line with findings in adults with ADPKD, where hypertension and cardiovascular damage precede decline in kidney function). We propose an algorithm for work-up and management based on current recommendations that integrates the need to screen regularly for hypertension and proteinuria in offspring of affected parents with different options regarding diagnostic testing, which need to be discussed with the family with regard to ethical and practical aspects. Indications and scope of genetic testing are discussed. Pharmacological management includes renin-angiotensin system blockade as first-line therapy for hypertension and proteinuria. The vasopressin receptor antagonist tolvaptan is licensed for delaying disease progression in adults with ADPKD who are likely to experience kidney failure. A clinical trial in children is currently ongoing; however, valid prediction models to identify children likely to suffer kidney failure are lacking. Non-pharmacological interventions in this population also deserve further study.
Collapse
|
22
|
Liebau MC, Mekahli D. Translational research approaches to study pediatric polycystic kidney disease. Mol Cell Pediatr 2021; 8:20. [PMID: 34882278 PMCID: PMC8660924 DOI: 10.1186/s40348-021-00131-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Accepted: 11/22/2021] [Indexed: 11/26/2022] Open
Abstract
Polycystic kidney diseases (PKD) are severe forms of genetic kidney disorders. The two main types of PKD are autosomal recessive and autosomal dominant PKD (ARPKD, ADPKD). While ARPKD typically is a disorder of early childhood, patients with ADPKD often remain pauci-symptomatic until adulthood even though formation of cysts in the kidney already begins in children. There is clinical and genetic overlap between both entities with very variable clinical courses. Subgroups of very early onset ADPKD may for example clinically resemble ARPKD. The basis of the clinical variability in both forms of PKD is not well understood and there are also limited prediction markers for disease progression for daily clinical life or surrogate endpoints for clinical trials in ARPKD or early ADPKD. As targeted therapeutic approaches to slow disease progression in PKD are emerging, it is becoming more important to reliably identify patients at risk for rapid progression as they might benefit from early therapy. Over the past years regional, national and international data collections to jointly analyze the clinical courses of PKD patients have been set up. The clinical observations are complemented by genetic studies and biorepositories as well as basic science approaches to elucidate the underlying molecular mechanisms in the PKD field. These approaches may serve as a basis for the development of novel therapeutic interventions in specific subgroups of patients. In this article we summarize some of the recent developments in the field with a focus on kidney involvement in PKD during childhood and adolescence and findings obtained in pediatric cohorts.
Collapse
Affiliation(s)
- Max Christoph Liebau
- Department of Pediatrics, Center for Rare Diseases and Center for Molecular Medicine, University Hospital Cologne and Medical Faculty, University of Cologne, Kerpener Str. 62, 50937, Cologne, Germany.
| | - Djalila Mekahli
- Department of Pediatric Nephrology and Organ Transplantation, University Hospitals Leuven, Herestraat 49, 3000, Leuven, Belgium. .,Department of Development and Regeneration, PKD Research Group, Laboratory of Pediatrics, KU Leuven, Leuven, Belgium.
| |
Collapse
|
23
|
Zhang Z, Bai H, Blumenfeld J, Ramnauth AB, Barash I, Prince M, Tan AY, Michaeel A, Liu G, Chicos I, Rennert L, Giannakopoulos S, Larbi K, Hughes S, Salvatore SP, Robinson BD, Kapur S, Rennert H. Detection of PKD1 and PKD2 Somatic Variants in Autosomal Dominant Polycystic Kidney Cyst Epithelial Cells by Whole-Genome Sequencing. J Am Soc Nephrol 2021; 32:3114-3129. [PMID: 34716216 PMCID: PMC8638386 DOI: 10.1681/asn.2021050690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/03/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder characterized by the development of multiple cysts in the kidneys. It is often caused by pathogenic mutations in PKD1 and PKD2 genes that encode polycystin proteins. Although the molecular mechanisms for cystogenesis are not established, concurrent inactivating germline and somatic mutations in PKD1 and PKD2 have been previously observed in renal tubular epithelium (RTE). METHODS To further investigate the cellular recessive mechanism of cystogenesis in RTE, we conducted whole-genome DNA sequencing analysis to identify germline variants and somatic alterations in RTE of 90 unique kidney cysts obtained during nephrectomy from 24 unrelated participants. RESULTS Kidney cysts were overall genomically stable, with low burdens of somatic short mutations or large-scale structural alterations. Pathogenic somatic "second hit" alterations disrupting PKD1 or PKD2 were identified in 93% of the cysts. Of these, 77% of cysts acquired short mutations in PKD1 or PKD2 ; specifically, 60% resulted in protein truncations (nonsense, frameshift, or splice site) and 17% caused non-truncating mutations (missense, in-frame insertions, or deletions). Another 18% of cysts acquired somatic chromosomal loss of heterozygosity (LOH) events encompassing PKD1 or PKD2 ranging from 2.6 to 81.3 Mb. 14% of these cysts harbored copy number neutral LOH events, while the other 3% had hemizygous chromosomal deletions. LOH events frequently occurred at chromosomal fragile sites, or in regions comprising chromosome microdeletion diseases/syndromes. Almost all somatic "second hit" alterations occurred at the same germline mutated PKD1/2 gene. CONCLUSIONS These findings further support a cellular recessive mechanism for cystogenesis in ADPKD primarily caused by inactivating germline and somatic variants of PKD1 or PKD2 genes in kidney cyst epithelium.
Collapse
Affiliation(s)
- Zhengmao Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Hanwen Bai
- Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Jon Blumenfeld
- Department of Medicine, Weill Cornell Medicine, New York, New York,The Rogosin Institute, New York, New York
| | - Andrew B. Ramnauth
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Irina Barash
- Department of Medicine, Weill Cornell Medicine, New York, New York,The Rogosin Institute, New York, New York
| | - Martin Prince
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Adrian Y. Tan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York,Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Alber Michaeel
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Genyan Liu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | - Lior Rennert
- Department of Public Health Sciences, Clemson University, Clemson, South Carolina
| | | | - Karen Larbi
- Vertex Pharmaceuticals Inc., Oxford, United Kingdom
| | | | - Steven P. Salvatore
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Sandip Kapur
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Hanna Rennert
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
24
|
Liebau MC. Is There a Functional Role of Mitochondrial Dysfunction in the Pathogenesis of ARPKD? Front Med (Lausanne) 2021; 8:739534. [PMID: 34676227 PMCID: PMC8523777 DOI: 10.3389/fmed.2021.739534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 09/03/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Max Christoph Liebau
- Department of Pediatrics, Center for Molecular Medicine, and Center for Rare Diseases, University Hospital Cologne and Medical Faculty, University of Cologne, Cologne, Germany
| |
Collapse
|
25
|
Liu F, Feng C, Shen H, Fu H, Mao J. Tolvaptan in Pediatric Autosomal Dominant Polycystic Kidney Disease: From Here to Where? KIDNEY DISEASES 2021; 7:343-349. [PMID: 34604341 DOI: 10.1159/000517186] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/08/2021] [Indexed: 12/17/2022]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disorder, accounting for approximately 5% of all ESRD cases worldwide. As a vasopressin receptor 2 antagonist, tolvaptan is the FDA-approved therapeutic agent for ADPKD, which is only made available to a limited number of adult patients; however, its efficacy in pediatric patients has not been reported widely. Summary Tolvaptan was shown to delay ADPKD progression in the Tolvaptan Efficacy and Safety in Management of Autosomal Dominant Polycystic Kidney Disease and Its Outcomes (TEMPO) 3:4 study, Replicating Evidence of Preserved Renal Function: an Investigation of Tolvaptan Safety and Efficacy in ADPKD (REPRISE) trial, and other clinical studies. In addition to its effects on aquaretic adverse events and alanine aminotransferase elevation, the effect of tolvaptan on ADPKD is clear, sustained, and cumulative. While ADPKD is a progressive disease, the early intervention has been shown to be important and beneficial in hypotheses as well as in trials. The use of tolvaptan in pediatric ADPKD involves the following challenges: patient assessment, quality of life assessment, cost-effectiveness, safety, and tolerability. The ongoing, phase 3b, 2-part study (ClinicalTrials.gov identifier: NCT02964273) on the evaluation of tolvaptan in pediatric ADPKD (patients aged 12-17 years) may help obtain some insights. Key Messages This review focuses on the rationality of tolvaptan use in pediatric patients with ADPKD, the associated challenges, and the suggested therapeutic approaches.
Collapse
Affiliation(s)
- Fei Liu
- Department of Nephrology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunyue Feng
- Department of Nephrology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huijun Shen
- Department of Nephrology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huaidong Fu
- Department of Nephrology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianhua Mao
- Department of Nephrology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Pinto E Vairo F, Prochnow C, Kemppainen JL, Lisi EC, Steyermark JM, Kruisselbrink TM, Pichurin PN, Dhamija R, Hager MM, Albadri S, Cornell LD, Lazaridis KN, Klee EW, Senum SR, El Ters M, Amer H, Baudhuin LM, Moyer AM, Keddis MT, Zand L, Sas DJ, Erickson SB, Fervenza FC, Lieske JC, Harris PC, Hogan MC. Genomics Integration Into Nephrology Practice. Kidney Med 2021; 3:785-798. [PMID: 34746741 PMCID: PMC8551494 DOI: 10.1016/j.xkme.2021.04.014] [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] [Indexed: 12/17/2022] Open
Abstract
RATIONALE & OBJECTIVE The etiology of kidney disease remains unknown in many individuals with chronic kidney disease (CKD). We created the Mayo Clinic Nephrology Genomics Clinic to improve our ability to integrate genomic and clinical data to identify the etiology of unexplained CKD. STUDY DESIGN Retrospective study. SETTING & PARTICIPANTS An essential component of our program is the Nephrology Genomics Board which consists of nephrologists, geneticists, pathologists, translational omics scientists, and trainees who interpret the patient's clinical and genetic data. Since September 2016, the Board has reviewed 163 cases (15 cystic, 100 glomerular, 6 congenital anomalies of kidney and urinary tract (CAKUT), 20 stones, 15 tubulointerstitial, and 13 other). ANALYTICAL APPROACH Testing was performed with targeted panels, single gene analysis, or analysis of kidney-related genes from exome sequencing. Variant classification was obtained based on the 2015 American College of Medical Genetics and Genomics and the Association for Molecular Pathology guidelines. RESULTS A definitive genetic diagnosis was achieved for 50 families (30.7%). The highest diagnostic yield was obtained in individuals with tubulointerstitial diseases (53.3%), followed by congenital anomalies of the kidney and urological tract (33.3%), glomerular (31%), cysts (26.7%), stones (25%), and others (15.4%). A further 20 (12.3%) patients had variants of interest, and variant segregation, and research activities (exome, genome, or transcriptome sequencing) are ongoing for 44 (40%) unresolved families. LIMITATIONS Possible overestimation of diagnostic rate due to inclusion of individuals with variants with evidence of pathogenicity but classified as of uncertain significance by the clinical laboratory. CONCLUSIONS Integration of genomic and research testing and multidisciplinary evaluation in a nephrology cohort with CKD of unknown etiology or suspected monogenic disease provided a diagnosis in a third of families. These diagnoses had prognostic implications, and often changes in management were implemented.
Collapse
Affiliation(s)
- Filippo Pinto E Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Carri Prochnow
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
| | | | - Emily C Lisi
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | - Joan M Steyermark
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Pavel N Pichurin
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
| | - Rhadika Dhamija
- Department of Clinical Genomics, Mayo Clinic, Scottsdale, Arizona
| | - Megan M Hager
- Department of Clinical Genomics, Mayo Clinic, Scottsdale, Arizona
| | - Sam Albadri
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota
| | - Lynn D Cornell
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota
| | - Konstantinos N Lazaridis
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Eric W Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Sarah R Senum
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Mireille El Ters
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hatem Amer
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Linnea M Baudhuin
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota
| | - Ann M Moyer
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota
| | - Mira T Keddis
- Division of Nephrology, Mayo Clinic, Scottsdale, Arizona
| | - Ladan Zand
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - David J Sas
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Stephen B Erickson
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - John C Lieske
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Laboratory Medicine & Pathology, Mayo Clinic, Rochester, Minnesota
| | - Peter C Harris
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Marie C Hogan
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Division of Nephrology & Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
27
|
Finnegan C, Murphy C, Breathnach F. Neonatal polycystic kidney disease: a novel variant. BMJ Case Rep 2021; 14:14/7/e242991. [PMID: 34290017 DOI: 10.1136/bcr-2021-242991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
Polycystic kidney disease (PKD) is a condition typified by multiple renal cysts and renal enlargement. Classification is usually determined by mode of inheritance-autosomal dominant PKD (ADPKD) or autosomal recessive PKD (ARPKD). ARPKD frequently presents in fetal life, but here we report a rare case of a family with two siblings diagnosed with ADPKD manifesting in utero with novel genetic findings. During the first pregnancy, enlarged cystic kidneys were noted at the gestational age (GA) of 18 weeks, which became progressively larger and anyhdramnios ensued by GA of 25 weeks. The couple opted to terminate the pregnancy. The second pregnancy similarly presented with bilateral enlarged cystic kidneys, but amniotic fluid remained normal throughout and she delivered at GA of 36 weeks. Genetic testing revealed the fetus to be heterozygous in AD PKD1, which is known to cause ADPKD and heterozygous for a hypomorphic allele for ADPKD of uncertain significance. The fetus was also found to be heterozygous in the AR PKHD1 gene with a variant not previously described in the literature. Where fetal features consistent with ARPKD are identified in the setting of familial ADPKD, this fetal manifestation of ADPKD, resulting from combined variants in the PKD1 gene, should be considered.
Collapse
Affiliation(s)
- Catherine Finnegan
- Obstetrics and Gynaecology, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Claire Murphy
- Departement of Paediatrics, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Fionnuala Breathnach
- Obstetrics and Gynaecology, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
28
|
Andolfo I, Martone S, Rosato BE, Marra R, Gambale A, Forni GL, Pinto V, Göransson M, Papadopoulou V, Gavillet M, Elalfy M, Panarelli A, Tomaiuolo G, Iolascon A, Russo R. Complex Modes of Inheritance in Hereditary Red Blood Cell Disorders: A Case Series Study of 155 Patients. Genes (Basel) 2021; 12:genes12070958. [PMID: 34201899 PMCID: PMC8304671 DOI: 10.3390/genes12070958] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/17/2021] [Accepted: 06/19/2021] [Indexed: 12/19/2022] Open
Abstract
Hereditary erythrocytes disorders include a large group of conditions with heterogeneous molecular bases and phenotypes. We analyzed here a case series of 155 consecutive patients with clinical suspicion of hereditary erythrocyte defects referred to the Medical Genetics Unit from 2018 to 2020. All of the cases followed a diagnostic workflow based on a targeted next-generation sequencing panel of 86 genes causative of hereditary red blood cell defects. We obtained an overall diagnostic yield of 84% of the tested patients. Monogenic inheritance was seen for 69% (107/155), and multi-locus inheritance for 15% (23/155). PIEZO1 and SPTA1 were the most mutated loci. Accordingly, 16/23 patients with multi-locus inheritance showed dual molecular diagnosis of dehydrated hereditary stomatocytosis/xerocytosis and hereditary spherocytosis. These dual inheritance cases were fully characterized and were clinically indistinguishable from patients with hereditary spherocytosis. Additionally, their ektacytometry curves highlighted alterations of dual inheritance patients compared to both dehydrated hereditary stomatocytosis and hereditary spherocytosis. Our findings expand the genotypic spectrum of red blood cell disorders and indicate that multi-locus inheritance should be considered for analysis and counseling of these patients. Of note, the genetic testing was crucial for diagnosis of patients with a complex mode of inheritance.
Collapse
Affiliation(s)
- Immacolata Andolfo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy; (I.A.); (S.M.); (B.E.R.); (R.M.); (R.R.)
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
| | - Stefania Martone
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy; (I.A.); (S.M.); (B.E.R.); (R.M.); (R.R.)
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
| | - Barbara Eleni Rosato
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy; (I.A.); (S.M.); (B.E.R.); (R.M.); (R.R.)
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
| | - Roberta Marra
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy; (I.A.); (S.M.); (B.E.R.); (R.M.); (R.R.)
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
| | - Antonella Gambale
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
- Department of Laboratory Medicine (DAIMedLab), UOC Medical Genetics, ‘Federico II’ University Hospital, 80131 Naples, Italy
| | - Gian Luca Forni
- Centro della Microcitemia e delle Anemie Congenite, Ospedale Galliera, 16128 Genoa, Italy; (G.L.F.); (V.P.)
| | - Valeria Pinto
- Centro della Microcitemia e delle Anemie Congenite, Ospedale Galliera, 16128 Genoa, Italy; (G.L.F.); (V.P.)
| | - Magnus Göransson
- Department of Paediatrics, The Queen Silvia Children’s Hospital, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden;
| | - Vasiliki Papadopoulou
- Service and Central Laboratory of Haematology, Department of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (V.P.); (M.G.)
| | - Mathilde Gavillet
- Service and Central Laboratory of Haematology, Department of Oncology and Department of Laboratory Medicine and Pathology, Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland; (V.P.); (M.G.)
| | - Mohsen Elalfy
- Thalassemia Centre, Faculty of Medicine, Ain Shams University, Cairo 11566, Egypt;
| | | | - Giovanna Tomaiuolo
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
- Department of Chemical Engineering, Materials and Industrial Production, ‘Federico II’ University of Naples, 80125 Naples, Italy
| | - Achille Iolascon
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy; (I.A.); (S.M.); (B.E.R.); (R.M.); (R.R.)
- CEINGE Biotecnologie Avanzate, 80145 Naples, Italy; (A.G.); (A.P.); (G.T.)
- Correspondence:
| | - Roberta Russo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli Federico II, 80131 Napoli, Italy; (I.A.); (S.M.); (B.E.R.); (R.M.); (R.R.)
| |
Collapse
|
29
|
Strong A, Simone L, Krentz A, Vaccaro C, Watson D, Ron H, Kalish JM, Pedro HF, Zackai EH, Hakonarson H. Expanding the genetic landscape of oral-facial-digital syndrome with two novel genes. Am J Med Genet A 2021; 185:2409-2416. [PMID: 34132027 PMCID: PMC8361718 DOI: 10.1002/ajmg.a.62337] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 12/18/2022]
Abstract
Oral‐facial‐digital syndromes (OFDS) are a heterogeneous and rare group of Mendelian disorders characterized by developmental abnormalities of the oral cavity, face, and digits caused by dysfunction of the primary cilium, a mechanosensory organelle that exists atop most cell types that facilitates organ patterning and growth. OFDS is inherited both in an X‐linked dominant, X‐linked recessive, and autosomal recessive manner. Importantly, though many of the causal genes for OFDS have been identified, up to 40% of OFD syndromes are of unknown genetic basis. Here we describe three children with classical presentations of OFDS including lingual hamartomas, polydactyly, and characteristic facial features found by exome sequencing to harbor variants in causal genes not previously associated with OFDS. We describe a female with hypothalamic hamartoma, urogenital sinus, polysyndactyly, and multiple lingual hamartomas consistent with OFDVI with biallelic pathogenic variants in CEP164, a gene associated with ciliopathy‐spectrum disease, but never before with OFDS. We additionally describe two unrelated probands with postaxial polydactyly, multiple lingual hamartomas, and dysmorphic features both found to be homozygous for an identical TOPORS missense variant, c.29 C>A; (p.Pro10Gln). Heterozygous TOPORS pathogenic gene variants are associated with autosomal dominant retinitis pigmentosa, but never before with syndromic ciliopathy. Of note, both probands are of Dominican ancestry, suggesting a possible founder allele.
Collapse
Affiliation(s)
- Alanna Strong
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Laurie Simone
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | | | - Courtney Vaccaro
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Deborah Watson
- The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hayley Ron
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jennifer M Kalish
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helio F Pedro
- Center for Genetic and Genomic Medicine, Hackensack University Medical Center, Hackensack, New Jersey, USA
| | - Elaine H Zackai
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hakon Hakonarson
- Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,The Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Division of Pulmonary Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
30
|
Benson KA, Murray SL, Senum SR, Elhassan E, Conlon ET, Kennedy C, Conlon S, Gilbert E, Connaughton D, O'Hara P, Khamis S, Cormican S, Brody LC, Molloy AM, Lynch SA, Casserly L, Griffin MD, Carton R, Yachnin K, Harris PC, Cavalleri GL, Conlon P. The genetic landscape of polycystic kidney disease in Ireland. Eur J Hum Genet 2021; 29:827-838. [PMID: 33454723 PMCID: PMC8110806 DOI: 10.1038/s41431-020-00806-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 12/16/2020] [Accepted: 12/21/2020] [Indexed: 02/08/2023] Open
Abstract
Polycystic kidney diseases (PKDs) comprise the most common Mendelian forms of renal disease. It is characterised by the development of fluid-filled renal cysts, causing progressive loss of kidney function, culminating in the need for renal replacement therapy or kidney transplant. Ireland represents a valuable region for the genetic study of PKD, as family sizes are traditionally large and the population relatively homogenous. Studying a cohort of 169 patients, we describe the genetic landscape of PKD in Ireland for the first time, compare the clinical features of patients with and without a molecular diagnosis and correlate disease severity with autosomal dominant pathogenic variant type. Using a combination of molecular genetic tools, including targeted next-generation sequencing, we report diagnostic rates of 71-83% in Irish PKD patients, depending on which variant classification guidelines are used (ACMG or Mayo clinic respectively). We have catalogued a spectrum of Irish autosomal dominant PKD pathogenic variants including 36 novel variants. We illustrate how apparently unrelated individuals carrying the same autosomal dominant pathogenic variant are highly likely to have inherited that variant from a common ancestor. We highlight issues surrounding the implementation of the ACMG guidelines for variant pathogenicity interpretation in PKD, which have important implications for clinical genetics.
Collapse
Affiliation(s)
- Katherine A Benson
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Susan L Murray
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland
| | - Sarah R Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | | | - Eoin T Conlon
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland
| | - Claire Kennedy
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland
| | - Shane Conlon
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland
| | - Edmund Gilbert
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Paul O'Hara
- Department of Renal Medicine, University Hospital Limerick, Limerick, Ireland
| | - Sarah Khamis
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Sarah Cormican
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| | - Lawrence C Brody
- Division of Intramural Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Anne M Molloy
- School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Sally Ann Lynch
- Department of Clinical Genetics, Children's University Hospital, Temple Street, Dublin, Ireland
| | - Liam Casserly
- Department of Renal Medicine, University Hospital Limerick, Limerick, Ireland
| | - Matthew D Griffin
- Regenerative Medicine Institute (REMEDI) at CÚRAM Centre for Research in Medical Devices, School of Medicine, National University of Ireland Galway, Galway, Ireland
- Nephrology Department, Galway University Hospitals, Saolta University Healthcare Group, Galway, Ireland
| | - Robert Carton
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Gianpiero L Cavalleri
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Peter Conlon
- School of Pharmacy and Biomolecular Science, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Department of Nephrology, Beaumont Hospital, Dublin, Ireland.
| |
Collapse
|
31
|
Arroyo J, Escobar-Zarate D, Wells HH, Constans MM, Thao K, Smith JM, Sieben CJ, Martell MR, Kline TL, Irazabal MV, Torres VE, Hopp K, Harris PC. The genetic background significantly impacts the severity of kidney cystic disease in the Pkd1 RC/RC mouse model of autosomal dominant polycystic kidney disease. Kidney Int 2021; 99:1392-1407. [PMID: 33705824 DOI: 10.1016/j.kint.2021.01.028] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/19/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), primarily due to PKD1 or PKD2 mutations, causes progressive kidney cyst development and kidney failure. There is significant intrafamilial variability likely due to the genetic background and environmental/lifestyle factors; variability that can be modeled in PKD mice. Here, we characterized mice homozygous for the PKD1 hypomorphic allele, p.Arg3277Cys (Pkd1RC/RC), inbred into the BALB/cJ (BC) or the 129S6/SvEvTac (129) strains, plus F1 progeny bred with the previously characterized C57BL/6J (B6) model; F1(BC/B6) or F1(129/B6). By one-month cystic disease in both the BC and 129 Pkd1RC/RC mice was more severe than in B6 and continued with more rapid progression to six to nine months. Thereafter, the expansive disease stage plateaued/declined, coinciding with increased fibrosis and a clear decline in kidney function. Greater severity correlated with more inter-animal and inter-kidney disease variability, especially in the 129-line. Both F1 combinations had intermediate disease severity, more similar to B6 but progressive from one-month of age. Mild biliary dysgenesis, and an early switch from proximal tubule to collecting duct cysts, was seen in all backgrounds. Preclinical testing with a positive control, tolvaptan, employed the F1(129/B6)-Pkd1RC/RC line, which has moderately progressive disease and limited isogenic variability. Magnetic resonance imaging was utilized to randomize animals and provide total kidney volume endpoints; complementing more traditional data. Thus, we show how genetic background can tailor the Pkd1RC/RC model to address different aspects of pathogenesis and disease modification, and describe a possible standardized protocol for preclinical testing.
Collapse
Affiliation(s)
- Jennifer Arroyo
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Harrison H Wells
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Megan M Constans
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Ka Thao
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Jessica M Smith
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Cynthia J Sieben
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Madeline R Martell
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Timothy L Kline
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Maria V Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Katharina Hopp
- Division of Renal Diseases and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, Colorado, USA.
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|