1
|
Tollaksen R, Craver RD, Yosypiv IV. Nephrotic Syndrome in a Child With NPHS2 Mutation. Pediatr Dev Pathol 2024; 27:359-363. [PMID: 38291869 DOI: 10.1177/10935266231223274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Steroid resistant nephrotic syndrome (SRNS) accounts for 30% of all cases of nephrotic syndrome (NS) in children and frequently leads to end stage kidney disease (ESKD). About 30% of children with SRNS demonstrate causative mutations in podocyte- associated genes. Early identification of genetic forms of SRNS is critical to avoid potentially harmful immunosuppressive therapy. A 2-year-old male patient with NS and no family history of renal disease did not respond to 4-week steroid treatment. Kidney biopsy demonstrated mesangial proliferative glomerulopathy with basement membrane dysmorphism. Tacrolimus and Lisinopril were added to therapy pending results of genetic testing. Kidney Gene panel showed a NPHS2 c.413G>A (p.Arg138Gln) homozygous pathogenic variant. This missense variant is considered a common pathogenic founder mutation in European populations. A diagnosis of autosomal-recessive form of nonsyndromic SRNS due to NPHS2 causative variant was made. Immunosuppresive therapy was stopped, Lizinopril dose was increased and weekly infusions of Albumin/furosemide were initiated to manage edema. This case demonstrates that early genetic testing in children with SRNS avoids prolonged potentially harmful immunosuppressive therapy, allows for timely genetic family counseling, and allows earlier consideration for future living related donor kidney transplantation.
Collapse
Affiliation(s)
- Ross Tollaksen
- Departments of Pediatrics, Section of Pediatric Nephrology, Tulane University Health Sciences Center, New Orleans, LA, USA
| | - Randall D Craver
- Department of Pathology, LSU School of Medicine, New Orleans, LA, USA
| | - Ihor V Yosypiv
- Departments of Pediatrics, Section of Pediatric Nephrology, Tulane University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
2
|
Mallawaarachchi AC, Fowles L, Wardrop L, Wood A, O'Shea R, Biros E, Harris T, Alexander SI, Bodek S, Boudville N, Burke J, Burnett L, Casauria S, Chadban S, Chakera A, Crafter S, Dai P, De Fazio P, Faull R, Honda A, Huntley V, Jahan S, Jayasinghe K, Jose M, Leaver A, MacShane M, Madelli EO, Nicholls K, Pawlowski R, Rangan G, Snelling P, Soraru J, Sundaram M, Tchan M, Valente G, Wallis M, Wedd L, Welland M, Whitlam J, Wilkins EJ, McCarthy H, Simons C, Quinlan C, Patel C, Stark Z, Mallett AJ. Genomic Testing in Patients with Kidney Failure of an Unknown Cause: A National Australian Study. Clin J Am Soc Nephrol 2024; 19:887-897. [PMID: 38861662 PMCID: PMC11254024 DOI: 10.2215/cjn.0000000000000464] [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: 10/30/2023] [Accepted: 04/25/2024] [Indexed: 06/13/2024]
Abstract
Key Points Twenty-five percent of those with unexplained kidney failure have a monogenic cause. Whole genome sequencing with broad gene panel analysis is a feasible diagnostic approach in nephrology. Background The cause of kidney failure is unknown in approximately 10% of patients with stage 5 chronic kidney disease (CKD). For those who first present to nephrology care with kidney failure, standard investigations of serology, imaging, urinalysis, and kidney biopsy are limited differentiators of etiology. We aimed to determine the diagnostic utility of whole genome sequencing (WGS) with analysis of a broad kidney gene panel in patients with kidney failure of unknown cause. Methods We prospectively recruited 100 participants who reached CKD stage 5 at the age of ≤50 years and had an unknown cause of kidney failure after standard investigation. Clinically accredited WGS was performed in this national cohort after genetic counseling. The primary analysis was targeted to 388 kidney-related genes with second-tier, genome-wide, and mitochondrial analysis. Results The cohort was 61% male and the average age of participants at stage 5 CKD was 32 years (9 months to 50 years). A genetic diagnosis was made in 25% of participants. Disease-causing variants were identified across autosomal dominant tubulointerstitial kidney disease (6), glomerular disorders (4), ciliopathies (3), tubular disorders (2), Alport syndrome (4), and mitochondrial disease (1). Most diagnoses (80%) were in autosomal dominant, X-linked, or mitochondrial conditions (UMOD ; COL4A5 ; INF2 ; CLCN5 ; TRPC6 ; COL4A4 ; EYA1 ; HNF1B ; WT1 ; NBEA ; m.3243A>G ). Participants with a family history of CKD were more likely to have a positive result (odds ratio, 3.29; 95% confidence interval, 1.10 to 11.29). Thirteen percent of participants without a CKD family history had a positive result. In those who first presented in stage 5 CKD, WGS with broad analysis of a curated kidney disease gene panel was diagnostically more informative than kidney biopsy, with biopsy being inconclusive in 24 of the 25 participants. Conclusions In this prospectively ascertained Australian cohort, we identified a genetic diagnosis in 25% of patients with kidney failure of unknown cause.
Collapse
Affiliation(s)
- Amali C. Mallawaarachchi
- Clinical Genetics Service, Institute of Precision Medicine and Bioinformatics, Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Genomic and Inherited Diseases Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- KidGen Collaborative, Australian Genomics Health Alliance, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Lindsay Fowles
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Louise Wardrop
- KidGen Collaborative, Kidney Regeneration, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Alasdair Wood
- KidGen Collaborative, Kidney Regeneration, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Rosie O'Shea
- KidGen Collaborative, Australian Genomics Health Alliance, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Erik Biros
- KidGen Collaborative, Australian Genomics Health Alliance, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- College of Medicine and Dentistry, James Cook University, Townsville, Queensland, Australia
- Townsville University Hospital, Townsville, Queensland, Australia
| | - Trudie Harris
- KidGen Collaborative, Australian Genomics Health Alliance, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Townsville University Hospital, Townsville, Queensland, Australia
| | - Stephen I. Alexander
- Centre for Kidney Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Department of Nephrology, Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Simon Bodek
- Clinical Genetics Service, Austin Health, Melbourne, Victoria, Australia
| | - Neil Boudville
- Medical School, University of Western Australia, Crawley, Western Australia, Australia
| | - Jo Burke
- School of Medicine and Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- Tasmanian Clinical Genetics Service, Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Leslie Burnett
- Genomic and Inherited Diseases Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Northern Clinical School, University of Sydney, Sydney, New South Wales, Australia
- St Vincent's Healthcare Clinical Campus, UNSW Sydney, Sydney, New South Wales, Australia
| | - Sarah Casauria
- Australian Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Steve Chadban
- Renal Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Aron Chakera
- Harry Perkins Institute for Medical Research, University of Western Australia, Crawley, Western Australia, Australia
- Renal Unit, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Sam Crafter
- The Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Pei Dai
- Precision Immunology Program, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, New South Wales, Australia
| | - Paul De Fazio
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Randall Faull
- Renal Unit, Royal Adelaide Hospital, Adelaide, South Australia, Australia
- University of Adelaide, Adelaide, South Australia, Australia
| | - Andrew Honda
- The Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Vanessa Huntley
- Adult Genetics Service, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Sadia Jahan
- The Central and Northern Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, South Australia, Australia
| | - Kushani Jayasinghe
- Department of Medicine, Monash University, Melbourne, Victoria, Australia
- Department of Nephrology, Monash Health, Melbourne, Victoria, Australia
- Melbourne Health, Melbourne, Victoria, Australia
| | - Matthew Jose
- Royal Hobart Hospital, Hobart, Tasmania, Australia
| | - Anna Leaver
- Clinical Genetics Service, Austin Health, Melbourne, Victoria, Australia
| | - Mandi MacShane
- Genetic Services of WA, KEMH, Subiaco, Western Australia, Australia
- Harry Perkins Institute of Medical Research, Nedlands, Western Australia, Australia
| | | | - Kathy Nicholls
- Nephrology Unit, Royal Melbourne Hospital, Parkville, Victoria, Australia
- The University of Melbourne, Parkville, Victoria, Australia
| | - Rhonda Pawlowski
- Anatomical Pathology, Monash Health, Melbourne, Victoria, Australia
| | - Gopi Rangan
- Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
- Michael Stern Laboratory for Polycystic Kidney Disease, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Paul Snelling
- Renal Medicine, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Jacqueline Soraru
- Department of Nephrology and Hypertension, Perth Children's Hospital, Nedlands, Western Australia, Australia
- Department of Nephrology and Renal Transplantation, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | | | - Michel Tchan
- Genetic Medicine, Westmead Hospital, Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Giulia Valente
- Clinical Genetics Service, Austin Health, Melbourne, Victoria, Australia
| | - Mathew Wallis
- School of Medicine and Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
- Tasmanian Clinical Genetics Service, Royal Hobart Hospital, Hobart, Tasmania, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Laura Wedd
- Centre for Population Genomics, Garvan Institute of Medical Research and UNSW Sydney, Sydney, New South Wales, Australia
| | - Matthew Welland
- Centre for Population Genomics, Garvan Institute of Medical Research and UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - John Whitlam
- Department of Nephrology, Austin Health, Melbourne, Victoria, Australia
| | - Ella J. Wilkins
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Hugh McCarthy
- Centre for Kidney Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Discipline of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Department of Nephrology, Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Cas Simons
- Centre for Population Genomics, Garvan Institute of Medical Research and UNSW Sydney, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Kidney Regeneration, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Catherine Quinlan
- Department of Kidney Regeneration, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Nephrology, Royal Children's Hospital, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Chirag Patel
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Zornitza Stark
- Australian Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J. Mallett
- KidGen Collaborative, Australian Genomics Health Alliance, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Townsville University Hospital, Townsville, Queensland, Australia
- Australian Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
- Institute for Molecular Bioscience and Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
3
|
Li X, Fan J, Fu R, Peng M, He J, Chen Q, Wang S, Chen C. Case report: A novel R246L mutation in the LMX1B homeodomain causes isolated nephropathy in a large Chinese family. Medicine (Baltimore) 2024; 103:e37442. [PMID: 38457557 PMCID: PMC10919518 DOI: 10.1097/md.0000000000037442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/09/2024] [Indexed: 03/10/2024] Open
Abstract
BACKGROUND Genetic factors contribute to chronic kidney disease (CKD) and end-stage renal disease (ESRD). Advances in genetic testing have enabled the identification of hereditary kidney diseases, including those caused by LMX1B mutations. LMX1B mutations can lead to nail-patella syndrome (NPS) or nail-patella-like renal disease (NPLRD) with only renal manifestations. CASE PRESENTATION The proband was a 13-year-old female who was diagnosed with nephrotic syndrome at the age of 6. Then she began intermittent hormone and drug therapy. When she was 13 years old, she was admitted to our hospital due to sudden chest tightness, which progressed to end-stage kidney disease (ESRD), requiring kidney replacement therapy. Whole-Exome Sequencing (WES) results suggest the presence of LMX1B gene mutation, c.737G > T, p.Arg246Leu. Tracing her family history, we found that her father, grandmother, uncle and 2 cousins all had hematuria, or proteinuria. In addition to the grandmother, a total of 9 members of the family performed WES. The members with kidney involved all carry the mutated gene. Healthy members did not have the mutated gene. It is characterized by co-segregation of genotype and phenotype. We followed the family for 9 year, the father developed ESRD at the age of 50 and started hemodialysis treatment. The rest patients had normal renal function. No extra-renal manifestations associated with NPS were found in any member of the family. CONCLUSIONS This study has successfully identified missense mutation, c.737G > T (p.Arg246Leu) in the homeodomain, which appears to be responsible for isolated nephropathy in the studied family. The arginine to leucine change at codon 246 likely disrupts the DNA-binding homeodomain of LMX1B. Previous research has documented 2 types of mutations at codon R246, namely R246Q and R246P, which are known to cause NPLRD. The newly discovered mutation, R246L, is likely to be another novel mutation associated with NPLRD, thus expanding the range of mutations at the crucial renal-critical codon 246 that contribute to the development of NPLRD. Furthermore, our findings suggest that any missense mutation occurring at the 246th amino acid position within the homeodomain of the LMX1B gene has the potential to lead to NPLRD.
Collapse
Affiliation(s)
- Xian Li
- Xinxiang Medical Univeisity, Xinxiang, China
- Puyang Oilfield General Hospital, Puyang, China
| | - Jiaojiao Fan
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rong Fu
- Puyang Oilfield General Hospital, Puyang, China
| | - Ming Peng
- Beijing Chigene Translational Medicine Research Center Co., Ltd, Beijing, China
| | - Jujie He
- Puyang Oilfield General Hospital, Puyang, China
| | | | | | - Chong Chen
- Puyang Oilfield General Hospital, Puyang, China
| |
Collapse
|
4
|
Thomasová D, Zelinová M, Libik M, Geryk J, Votýpka P, Rajnochová Bloudíčková S, Krejčí K, Reiterová J, Jančová E, Machová J, Kollárová M, Rychík I, Havrda M, Horáčková M, Putzová M, Šafránek R, Kollár M, Macek M. The most common founder pathogenic variant c.868G > A (p.Val290Met) in the NPHS2 gene in a representative adult Czech cohort with focal segmental glomerulosclerosis is associated with a milder disease and its underdiagnosis in childhood. Front Med (Lausanne) 2023; 10:1320054. [PMID: 38170106 PMCID: PMC10759319 DOI: 10.3389/fmed.2023.1320054] [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: 10/11/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Background Genetic focal segmental glomerulosclerosis (FSGS) is caused by pathogenic variants in a broad spectrum of genes that have a variable representation based on subjects' ethnicity and/or age. The most frequently mutated autosomal recessive gene in FSGS is NPHS2. In this study, we analyzed the spectrum of NPHS2 variants and their associated phenotype in Czech adult FSGS patients. Methods A representative cohort of 234 adult patients with FSGS, derived from 225 families originating from all regions of Czechia, was analyzed by massively parallel sequencing. In this study, we focused on the comprehensive analysis of the NPHS2 gene. The histological classification of FSGS followed the Columbia classification. Results We detected seven (3%) cases bearing homozygous or compound heterozygous pathogenic NPHS2 variants. A single pathogenic variant c.868G > A (p.Val290Met) was found in the majority of NPHS2-positive cases (86%; 6 out of 7) in histologically confirmed instances of FSGS. Its allele frequency among unrelated NPHS2-associated FSGS patients was 50% (6/12), and Haplotype analysis predicted its origin to be a result of a founder effect. There is an identical V290M-related haplotype on all V290M alleles spanning a 0,7 Mb region flanking NPHS2 in Central European FSGS populations. The phenotype of the p.Val290Met NPHS2-associated FSGS demonstrated a later onset and a much milder course of the disease compared to other NPHS2 pathogenic variants associated with FSGS. The mean age of the FSGS diagnosis based on kidney biopsy evaluation was 31.2 ± 7.46 years. In 50% of all cases, the initial disease manifestation of proteinuria occurred only in adulthood, with 83% of these cases not presenting with edemas. One-third (33%) of the studied subjects progressed to ESRD (2 out of 6) at the mean age of 35.0 ± 2.82 years. Conclusions We identified the most prevalent pathogenic variant, p.Val290Met, in the NPHS2 gene among Czech adult FSGS patients, which has arisen due to a founder effect in Central Europe. The documented milder course of the disease associated with this variant leads to the underdiagnosis in childhood. We established the histopathological features of the NPHS2-associated adult FSGS cases based on the Columbia classification. This might improve patient stratification and optimize their treatment.
Collapse
Affiliation(s)
- Dana Thomasová
- Institute of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University Prague, Prague, Czechia
| | - Michaela Zelinová
- Institute of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University Prague, Prague, Czechia
| | - Malgorzata Libik
- Institute of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University Prague, Prague, Czechia
| | - Jan Geryk
- Institute of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University Prague, Prague, Czechia
| | - Pavel Votýpka
- Institute of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University Prague, Prague, Czechia
| | | | - Karel Krejčí
- 3rd Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital and Faculty of Medicine Palacký University Olomouc, Olomouc, Czechia
| | - Jana Reiterová
- Department of Nephrology, General University Hospital in Prague, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Eva Jančová
- Department of Nephrology, General University Hospital in Prague, First Faculty of Medicine, Charles University, Prague, Czechia
| | - Jana Machová
- Department of Internal Medicine I, Faculty of Medicine in Pilsen, Charles University and Teaching Hospital, Pilsen, Czechia
- Biomedical Centre, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czechia
| | - Martina Kollárová
- Department of Internal Medicine, University Hospital Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Ivan Rychík
- Department of Internal Medicine, University Hospital Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Martin Havrda
- Department of Internal Medicine, University Hospital Vinohrady and Third Faculty of Medicine, Charles University, Prague, Czechia
| | - Miroslava Horáčková
- Department of Internal Medicine, University Hospital Motol and 2nd Faculty of Medicine, Charles University, Prague, Czechia
| | - Martina Putzová
- Biopticka Laboratory, Pilsen, Czechia
- Faculty of Medicine in Plzeň - Charles University, Pilsen, Czechia
| | - Roman Šafránek
- Department of Nephrology, University Hospital Hradec Králové, Faculty of Medicine in Hradec Králové, Charles University, Hradec Králové, Czechia
| | - Marek Kollár
- Department of Pathology, Institute for Clinical and Experimental Medicine, Prague, Czechia
| | - Milan Macek
- Institute of Biology and Medical Genetics, University Hospital Motol and 2nd Faculty of Medicine, Charles University Prague, Prague, Czechia
| |
Collapse
|
5
|
Ding WY, Kuzmuk V, Hunter S, Lay A, Hayes B, Beesley M, Rollason R, Hurcombe JA, Barrington F, Masson C, Cathery W, May C, Tuffin J, Roberts T, Mollet G, Chu CJ, McIntosh J, Coward RJ, Antignac C, Nathwani A, Welsh GI, Saleem MA. Adeno-associated virus gene therapy prevents progression of kidney disease in genetic models of nephrotic syndrome. Sci Transl Med 2023; 15:eabc8226. [PMID: 37556557 DOI: 10.1126/scitranslmed.abc8226] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/21/2023] [Indexed: 08/11/2023]
Abstract
Gene therapy for kidney diseases has proven challenging. Adeno-associated virus (AAV) is used as a vector for gene therapy targeting other organs, with particular success demonstrated in monogenic diseases. We aimed to establish gene therapy for the kidney by targeting a monogenic disease of the kidney podocyte. The most common cause of childhood genetic nephrotic syndrome is mutations in the podocyte gene NPHS2, encoding podocin. We used AAV-based gene therapy to rescue this genetic defect in human and mouse models of disease. In vitro transduction studies identified the AAV-LK03 serotype as a highly efficient transducer of human podocytes. AAV-LK03-mediated transduction of podocin in mutant human podocytes resulted in functional rescue in vitro, and AAV 2/9-mediated gene transfer in both the inducible podocin knockout and knock-in mouse models resulted in successful amelioration of kidney disease. A prophylactic approach of AAV 2/9 gene transfer before induction of disease in conditional knockout mice demonstrated improvements in albuminuria, plasma creatinine, plasma urea, plasma cholesterol, histological changes, and long-term survival. A therapeutic approach of AAV 2/9 gene transfer 2 weeks after disease induction in proteinuric conditional knock-in mice demonstrated improvement in urinary albuminuria at days 42 and 56 after disease induction, with corresponding improvements in plasma albumin. Therefore, we have demonstrated successful AAV-mediated gene rescue in a monogenic renal disease and established the podocyte as a tractable target for gene therapy approaches.
Collapse
Affiliation(s)
- Wen Y Ding
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Valeryia Kuzmuk
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
- Purespring Therapeutics, Rolling Stock Yard, 188 York Way, London N7 9AS, UK
| | - Sarah Hunter
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Abigail Lay
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Bryony Hayes
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Matthew Beesley
- Department of Histopathology, Cheltenham General Hospital, Cheltenham GL53 7AN, UK
| | - Ruth Rollason
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Jennifer A Hurcombe
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Fern Barrington
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Catrin Masson
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - William Cathery
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Carl May
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Jack Tuffin
- Purespring Therapeutics, Rolling Stock Yard, 188 York Way, London N7 9AS, UK
| | - Timothy Roberts
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Geraldine Mollet
- Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Université Paris Cité, Paris 75015, France
| | - Colin J Chu
- Academic Unit of Ophthalmology, Bristol Medical School, Biomedical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Jenny McIntosh
- Research Department of Haematology, UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6BT, UK
| | - Richard J Coward
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Corinne Antignac
- Laboratoire des Maladies Rénales Héréditaires, Inserm UMR 1163, Institut Imagine, Université Paris Cité, Paris 75015, France
| | - Amit Nathwani
- Research Department of Haematology, UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6BT, UK
| | - Gavin I Welsh
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| | - Moin A Saleem
- Bristol Renal, Bristol Medical School, Dorothy Hodgkin Building, University of Bristol, Bristol BS1 3NY, UK
| |
Collapse
|
6
|
Genetic nephrotic syndrome associated with disturbed function of glomerular slit membrane and podocyte cytoskeleton in children. Clin Exp Nephrol 2023; 27:101-109. [PMID: 36482266 DOI: 10.1007/s10157-022-02305-x] [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: 10/05/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Genetic nephrotic syndrome is caused by pathogenic variants in genes encoding proteins necessary for the stability and functionality of the glomerular filtration barrier. To date, more than 70 genes associated with steroid-resistant nephrotic syndrome have been identified. We review the clinical and molecular aspects of genetic nephrotic syndrome with a particular focus on genes associated with slit membrane and podocyte cytoskeleton defects. Sanger sequencing and next-generation sequencing are widely used in the identification of novel gene variants and help us gain a better understanding of the disease. Despite these findings, therapy is mainly supportive and focused on the reduction of proteinuria and management of chronic kidney disease with an unfavorable outcome for a significant proportion of cases. Positive therapeutic effects of immunosuppressive drugs have been reported in some patients; however, their long-time administration cannot be generally recommended. CONCLUSION Personalized treatment based on understanding the distinct disease pathogenesis is needed. With this, it will be possible to avoid harmful immunosuppressive therapy and improve outcomes and quality of life for pediatric patients suffering from genetic nephrotic syndrome.
Collapse
|
7
|
Dorison A, Ghobrial I, Graham A, Peiris T, Forbes TA, See M, Das M, Saleem MA, Quinlan C, Lawlor KT, Ramialison M, Howden SE, Little MH. Kidney Organoids Generated Using an Allelic Series of NPHS2 Point Variants Reveal Distinct Intracellular Podocin Mistrafficking. J Am Soc Nephrol 2023; 34:88-109. [PMID: 36167728 PMCID: PMC10101587 DOI: 10.1681/asn.2022060707] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 08/31/2022] [Accepted: 09/06/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND NPHS2 variants are the most common cause of steroid-resistant nephrotic syndrome in children >1 month old. Missense NPHS2 variants were reported to cause mistrafficking of the encoded protein, PODOCIN, but this conclusion was on the basis of overexpression in some nonpodocyte cell lines. METHODS We generated a series of human induced pluripotent stem cell (iPSC) lines bearing pathogenic missense variants of NPHS2 , encoding the protein changes p.G92C, p.P118L, p.R138Q, p.R168H, and p.R291W, and control lines. iPSC lines were also generated from a patient with steroid-resistant nephrotic syndrome (p.R168H homozygote) and a healthy heterozygous parent. All lines were differentiated into kidney organoids. Immunofluorescence assessed PODOCIN expression and subcellular localization. Podocytes were transcriptionally profiled and PODOCIN-NEPHRIN interaction interrogated. RESULTS All variant lines revealed reduced levels of PODOCIN protein in the absence of reduced transcription. Although wild-type PODOCIN localized to the membrane, distinct variant proteins displayed unique patterns of subcellular protein trafficking, some unreported. P118L and R138Q were preferentially retained in the endoplasmic reticulum (ER); R168H and R291W accumulated in the Golgi. Podocyte profiling demonstrated minimal disease-associated transcriptional change. All variants displayed podocyte-specific apoptosis, which was not linked to ER stress. NEPHRIN-PODOCIN colocalization elucidated the variant-specific effect on NEPHRIN association and hence NEPHRIN trafficking. CONCLUSIONS Specific variants of endogenous NPHS2 result in distinct subcellular PODOCIN localization within organoid podocytes. Understanding the effect of each variant on protein levels and localization and the effect on NEPHRIN provides additional insight into the pathobiology of NPHS2 variants. PODCAST This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2023_01_05_JASN2022060707.mp3.
Collapse
Affiliation(s)
- Aude Dorison
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Irene Ghobrial
- Murdoch Children’s Research Institute, Melbourne, Australia
| | - Alison Graham
- Murdoch Children’s Research Institute, Melbourne, Australia
| | | | - Thomas A. Forbes
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Royal Children’s Hospital, Melbourne, Australia
| | - Michael See
- Murdoch Children’s Research Institute, Melbourne, Australia
- Monash Bioinformatics Platform, Monash University, Clayton, Australia
| | - Mithun Das
- Murdoch Children’s Research Institute, Melbourne, Australia
| | - Moin A. Saleem
- Department of Paediatric Nephrology, Bristol Royal Hospital for Children, Bristol, United Kingdom
| | - Catherine Quinlan
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Royal Children’s Hospital, Melbourne, Australia
| | - Kynan T. Lawlor
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Mirana Ramialison
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
- Australian Regenerative Medicine Institute, Clayton, Australia
| | - Sara E. Howden
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Melissa H. Little
- Murdoch Children’s Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
- Novo Nordisk Foundation Centre for Stem Cell Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Staruschenko A, Ma R, Palygin O, Dryer SE. Ion channels and channelopathies in glomeruli. Physiol Rev 2023; 103:787-854. [PMID: 36007181 PMCID: PMC9662803 DOI: 10.1152/physrev.00013.2022] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 08/15/2022] [Accepted: 08/21/2022] [Indexed: 11/22/2022] Open
Abstract
An essential step in renal function entails the formation of an ultrafiltrate that is delivered to the renal tubules for subsequent processing. This process, known as glomerular filtration, is controlled by intrinsic regulatory systems and by paracrine, neuronal, and endocrine signals that converge onto glomerular cells. In addition, the characteristics of glomerular fluid flow, such as the glomerular filtration rate and the glomerular filtration fraction, play an important role in determining blood flow to the rest of the kidney. Consequently, disease processes that initially affect glomeruli are the most likely to lead to end-stage kidney failure. The cells that comprise the glomerular filter, especially podocytes and mesangial cells, express many different types of ion channels that regulate intrinsic aspects of cell function and cellular responses to the local environment, such as changes in glomerular capillary pressure. Dysregulation of glomerular ion channels, such as changes in TRPC6, can lead to devastating glomerular diseases, and a number of channels, including TRPC6, TRPC5, and various ionotropic receptors, are promising targets for drug development. This review discusses glomerular structure and glomerular disease processes. It also describes the types of plasma membrane ion channels that have been identified in glomerular cells, the physiological and pathophysiological contexts in which they operate, and the pathways by which they are regulated and dysregulated. The contributions of these channels to glomerular disease processes, such as focal segmental glomerulosclerosis (FSGS) and diabetic nephropathy, as well as the development of drugs that target these channels are also discussed.
Collapse
Affiliation(s)
- Alexander Staruschenko
- Department of Molecular Pharmacology and Physiology, University of South Florida, Tampa, Florida
- Hypertension and Kidney Research Center, University of South Florida, Tampa, Florida
- James A. Haley Veterans Hospital, Tampa, Florida
| | - Rong Ma
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Oleg Palygin
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Stuart E Dryer
- Department of Biology and Biochemistry, University of Houston, Houston, Texas
- Department of Biomedical Sciences, Tilman J. Fertitta Family College of Medicine, University of Houston, Houston, Texas
| |
Collapse
|
9
|
Navarro-Betancourt JR, Cybulsky AV. The IRE1α pathway in glomerular diseases: The unfolded protein response and beyond. FRONTIERS IN MOLECULAR MEDICINE 2022; 2:971247. [PMID: 39086958 PMCID: PMC11285563 DOI: 10.3389/fmmed.2022.971247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 09/07/2022] [Indexed: 08/02/2024]
Abstract
Endoplasmic reticulum (ER) function is vital for protein homeostasis ("proteostasis"). Protein misfolding in the ER of podocytes (glomerular visceral epithelial cells) is an important contributor to the pathogenesis of human glomerular diseases. ER protein misfolding causes ER stress and activates a compensatory signaling network called the unfolded protein response (UPR). Disruption of the UPR, in particular deletion of the UPR transducer, inositol-requiring enzyme 1α (IRE1α) in mouse podocytes leads to podocyte injury and albuminuria in aging, and exacerbates injury in glomerulonephritis. The UPR may interact in a coordinated manner with autophagy to relieve protein misfolding and its consequences. Recent studies have identified novel downstream targets of IRE1α, which provide new mechanistic insights into proteostatic pathways. Novel pathways of IRE1α signaling involve reticulophagy, mitochondria, metabolism, vesicular trafficking, microRNAs, and others. Mechanism-based therapies for glomerulopathies are limited, and development of non-invasive ER stress biomarkers, as well as targeting ER stress with pharmacological compounds may represent a therapeutic opportunity for preventing or attenuating progression of chronic kidney disease.
Collapse
Affiliation(s)
| | - Andrey V. Cybulsky
- Department of Medicine, McGill University Health Centre Research Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
10
|
Jankowski M, Daca-Roszak P, Obracht-Prondzyński C, Płoski R, Lipska-Ziętkiewicz BS, Ziętkiewicz E. Genetic diversity in Kashubs: the regional increase in the frequency of several disease-causing variants. J Appl Genet 2022; 63:691-701. [PMID: 35971028 PMCID: PMC9637066 DOI: 10.1007/s13353-022-00713-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 12/02/2022]
Abstract
Differential distribution of genetic variants’ frequency among human populations is caused by the genetic drift in isolated populations, historical migrations, and demography. Some of these variants are identical by descent and represent founder mutations, which — if pathogenic in nature — lead to the increased frequency of otherwise rare diseases. The detection of the increased regional prevalence of pathogenic variants may shed light on the historical processes that affected studied populations and can help to develop effective screening and diagnostic strategies as a part of personalized medicine. Here, we discuss the specific genetic diversity in Kashubs, the minority group living in northern Poland, reflected in the biased distribution of some of the repetitively found disease-causing variants. These include the following: (1) c.662A > G (p.Asp221Gly) in LDLR, causing heterozygous familial hypercholesterolemia; (2) c.3700_3704del in BRCA1, associated with hereditary breast and ovarian cancer syndrome; (3) c.1528G > C (p.Glu510Gln) in HADHA, seen in long-chain 3-hydroxy acyl-CoA dehydrogenase (LCHAD) deficiency, and (4) c.1032delT in NPHS2, associated with steroid-resistant nephrotic syndrome.
Collapse
Affiliation(s)
- Maciej Jankowski
- Department of Biology and Medical Genetics, Medical University of Gdansk, Gdansk, Poland
| | | | | | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, Warsaw, Poland
| | - Beata S Lipska-Ziętkiewicz
- Clinical Genetics Unit, Department of Biology and Medical Genetics, Medical University of Gdansk, Gdansk, Poland. .,Centre for Rare Diseases, Medical University of Gdansk, Gdansk, Poland.
| | - Ewa Ziętkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland.
| |
Collapse
|
11
|
Dorval G, Servais A, Boyer O. The genetics of steroid-resistant nephrotic syndrome in children. Nephrol Dial Transplant 2022; 37:648-651. [DOI: 10.1093/ndt/gfaa221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Indexed: 11/14/2022] Open
Affiliation(s)
- Guillaume Dorval
- Service de génétique moléculaire, Hôpital Necker - Enfants Malades, APHP, Paris, France
- Inserm U1163, Institut Imagine, Université de Paris, Paris, France
| | - Aude Servais
- Inserm U1163, Institut Imagine, Université de Paris, Paris, France
- Néphrologie et Transplantation, centre de référence MARHEA, Hôpital Necker - Enfants Malades, APHP, Paris, France
| | - Olivia Boyer
- Inserm U1163, Institut Imagine, Université de Paris, Paris, France
- Néphrologie pédiatrique, centre de référence MARHEA, centre de référence du syndrome néphrotique idiopathique de l’enfant et de l’adulte, Hôpital Necker - Enfants Malades, APHP, Paris, France
| |
Collapse
|
12
|
Spectrum of NPHS1 and NPHS2 variants in egyptian children with focal segmental glomerular sclerosis: identification of six novel variants and founder effect. Mol Genet Genomics 2022; 297:689-698. [DOI: 10.1007/s00438-022-01877-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 02/20/2022] [Indexed: 01/10/2023]
|
13
|
Ma C, Wang C, Luo D, Yan L, Yang W, Li N, Gao N. Structural insights into the membrane microdomain organization by SPFH family proteins. Cell Res 2022; 32:176-189. [PMID: 34975153 PMCID: PMC8807802 DOI: 10.1038/s41422-021-00598-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 11/09/2021] [Indexed: 02/03/2023] Open
Abstract
The lateral segregation of membrane constituents into functional microdomains, conceptually known as lipid raft, is a universal organization principle for cellular membranes in both prokaryotes and eukaryotes. The widespread Stomatin, Prohibitin, Flotillin, and HflK/C (SPFH) family proteins are enriched in functional membrane microdomains at various subcellular locations, and therefore were hypothesized to play a scaffolding role in microdomain formation. In addition, many SPFH proteins are also implicated in highly specific processes occurring on the membrane. However, none of these functions is understood at the molecular level. Here we report the structure of a supramolecular complex that is isolated from bacterial membrane microdomains and contains two SPFH proteins (HflK and HflC) and a membrane-anchored AAA+ protease FtsH. HflK and HflC form a circular 24-mer assembly, featuring a laterally segregated membrane microdomain (20 nm in diameter) bordered by transmembrane domains of HflK/C and a completely sealed periplasmic vault. Four FtsH hexamers are embedded inside this microdomain through interactions with the inner surface of the vault. These observations provide a mechanistic explanation for the role of HflK/C and their mitochondrial homologs prohibitins in regulating membrane-bound AAA+ proteases, and suggest a general model for the organization and functionalization of membrane microdomains by SPFH proteins.
Collapse
Affiliation(s)
- Chengying Ma
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Chengkun Wang
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Dingyi Luo
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Lu Yan
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Wenxian Yang
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Ningning Li
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China
| | - Ning Gao
- grid.11135.370000 0001 2256 9319State Key Laboratory of Membrane Biology, Peking-Tsinghua Joint Centre for Life Sciences, School of Life Sciences, Peking University, Beijing, China ,grid.11135.370000 0001 2256 9319National Biomedical Imaging Center, Peking University, Beijing, China
| |
Collapse
|
14
|
Agarwal S, Sudhini YR, Polat OK, Reiser J, Altintas MM. Renal cell markers: lighthouses for managing renal diseases. Am J Physiol Renal Physiol 2021; 321:F715-F739. [PMID: 34632812 DOI: 10.1152/ajprenal.00182.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Kidneys, one of the vital organs in our body, are responsible for maintaining whole body homeostasis. The complexity of renal function (e.g., filtration, reabsorption, fluid and electrolyte regulation, and urine production) demands diversity not only at the level of cell types but also in their overall distribution and structural framework within the kidney. To gain an in depth molecular-level understanding of the renal system, it is imperative to discern the components of kidney and the types of cells residing in each of the subregions. Recent developments in labeling, tracing, and imaging techniques have enabled us to mark, monitor, and identify these cells in vivo with high efficiency in a minimally invasive manner. In this review, we summarize different cell types, specific markers that are uniquely associated with those cell types, and their distribution in the kidney, which altogether make kidneys so special and different. Cellular sorting based on the presence of certain proteins on the cell surface allowed for the assignment of multiple markers for each cell type. However, different studies using different techniques have found contradictions in cell type-specific markers. Thus, the term "cell marker" might be imprecise and suboptimal, leading to uncertainty when interpreting the data. Therefore, we strongly believe that there is an unmet need to define the best cell markers for a cell type. Although the compendium of renal-selective marker proteins presented in this review is a resource that may be useful to researchers, we acknowledge that the list may not be necessarily exhaustive.
Collapse
Affiliation(s)
- Shivangi Agarwal
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | | - Onur K Polat
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | - Jochen Reiser
- Department of Internal Medicine, Rush University, Chicago, Illinois
| | | |
Collapse
|
15
|
Urinary podocyte markers in kidney diseases. Clin Chim Acta 2021; 523:315-324. [PMID: 34666027 DOI: 10.1016/j.cca.2021.10.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/17/2021] [Accepted: 10/13/2021] [Indexed: 01/19/2023]
Abstract
Podocytes play an important role in the maintenance of kidney function, and they are the primary focus of many kidney diseases. Podocyte injury results in the shedding of podocyte-derived cellular fragments and podocyte-specific molecular targets into the urine, which may serve as biomarkers of kidney diseases. Intact podocytes, either viable or dead, and podocyte-derived microvesicles could be quantified in the urine by various centrifugation, visualization and culture methods. Podocyte-specific protein targets from the nucleus, cytoplasm, slit-diaphragm, glomerular capillary basement membrane, and cytoskeleton, as well as their corresponding messenger RNA (mRNA), in the urine could be quantified by western blotting, ELISA, or quantitative polymerase chain reaction. Although some of these techniques may be expensive or labor-intensive at present, they may become widely available in the future because of the improvement in technology and automation. The application of urinary podocyte markers for the diagnosis and monitoring of various kidney diseases have been explored but the published data in this area are not sufficiently systematic and lack external validation. Further research should focus on standardizing, comparing, and automizing laboratory methods, as well as defining their added value to the routine clinical tests.
Collapse
|
16
|
Schrezenmeier E, Kremerskothen E, Halleck F, Staeck O, Liefeldt L, Choi M, Schüler M, Weber U, Bachmann N, Grohmann M, Wagner T, Budde K, Bergmann C. The underestimated burden of monogenic kidney disease in adults waitlisted for kidney transplantation. Genet Med 2021; 23:1219-1224. [PMID: 33712733 PMCID: PMC8257480 DOI: 10.1038/s41436-021-01127-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose Chronic kidney disease (CKD) is a major health-care burden. Increasing evidence suggests that a considerable proportion of patients are affected by a monogenic kidney disorder. Methods In this study, the kidney transplantation waiting list at the Charité was screened for patients with undetermined cause of CKD. By next-generation sequencing (NGS) we targeted all 600 genes described and associated with kidney disease or allied disorders. Results In total, 635 patients were investigated. Of these, 245 individuals had a known cause of CKD (38.5%) of which 119 had a proven genetic disease (e.g., ADPKD, Alport). The other 340 patients (53.5%) were classified as undetermined diagnosis, of whom 87 had kidney failure (KF) onset <40 years. To this latter group genetic testing was offered as well as to those patients (n = 29) with focal segmental glomerulosclerosis (FSGS) and all individuals (n = 21) suspicious for thrombotic microangiopathy (TMA) in kidney biopsy. We detected diagnostic variants in 26 of 126 patients (20.6%) of which 14 of 126 (11.1%) were pathogenic or likely pathogenic. In another 12 of 126 (9.5%) patients, variants of unknown significance (VUS) were detected. Conclusion Our study demonstrates the diagnostic value of comprehensive genetic testing among patients with undetermined CKD.
Collapse
Affiliation(s)
- Eva Schrezenmeier
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany. .,Berlin Institute of Health (BIH), Berlin, Germany.
| | - Elisa Kremerskothen
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Fabian Halleck
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | | | - Lutz Liefeldt
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Mira Choi
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Markus Schüler
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Ulrike Weber
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Nadine Bachmann
- Medizinische Genetik Mainz, Limbach Genetics GmbH, Mainz, Germany
| | - Maik Grohmann
- Medizinische Genetik Mainz, Limbach Genetics GmbH, Mainz, Germany
| | - Timo Wagner
- Medizinische Genetik Mainz, Limbach Genetics GmbH, Mainz, Germany
| | - Klemens Budde
- Charité-Universitätsmedizin Berlin, Department of Nephrology and Medical Intensive Care, Berlin, Germany
| | - Carsten Bergmann
- Medizinische Genetik Mainz, Limbach Genetics GmbH, Mainz, Germany. .,Department of Medicine, Nephrology, University Hospital Freiburg, Freiburg, Germany.
| |
Collapse
|
17
|
Next-generation sequencing in patients with familial FSGS: first report of collagen gene mutations in Tunisian patients. J Hum Genet 2021; 66:795-803. [PMID: 33654185 DOI: 10.1038/s10038-021-00912-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/09/2021] [Accepted: 02/16/2021] [Indexed: 11/08/2022]
Abstract
Focal segmental glomerulosclerosis (FSGS) is a histological lesion with many causes, including inherited genetic defects, with significant proteinuria being the predominant clinical finding at presentation. FSGS is considered as a podocyte disease due to the fact that in the majority of patients with FSGS, the lesion results from defects in the podocyte structure. However, FSGS does not result exclusively from podocyte-associated genes. In this study, we used a genetic approach based on targeted next-generation sequencing (NGS) of 242 genes to identify the genetic cause of FSGS in seven Tunisian families. The sequencing results revealed the presence of eight distinct mutations including seven newly discovered ones: the c.538G>A (p.V180M) in NPHS2, c.5186G>A (p.R1729Q) in PLCE1 and c.232A>C (p.I78L) in PAX2 and five novel mutations in COL4A3 and COL4A4 genes. Four mutations (c.209G>A (p.G70D), c.725G>A (p.G242E), c.2225G>A (p.G742E), and c. 1681_1698del) were detected in COL4A3 gene and one mutation (c.1424G>A (p.G475D)) was found in COL4A4. In summary, NGS of a targeted gene panel is an ideal approach for the genetic testing of FSGS with multiple possible underlying etiologies. We have demonstrated that not only podocyte genes but also COL4A3/4 mutations should be considered in patients with FSGS.
Collapse
|
18
|
Tokhmafshan F, Dickinson K, Akpa MM, Brasell E, Huertas P, Goodyer PR. A no-nonsense approach to hereditary kidney disease. Pediatr Nephrol 2020; 35:2031-2042. [PMID: 31807928 DOI: 10.1007/s00467-019-04394-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/05/2019] [Accepted: 10/07/2019] [Indexed: 01/12/2023]
Abstract
The advent of a new class of aminoglycosides with increased translational readthrough of nonsense mutations and reduced toxicity offers a new therapeutic strategy for a subset of patients with hereditary kidney disease. The renal uptake and retention of aminoglycosides at a high intracellular concentration makes the kidney an ideal target for this approach. In this review, we explore the potential of aminoglycoside readthrough therapy in a number of hereditary kidney diseases and discuss the therapeutic window of opportunity for subclasses of each disease, when caused by nonsense mutations.
Collapse
Affiliation(s)
- Fatima Tokhmafshan
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada
| | - Kyle Dickinson
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada.,Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Murielle M Akpa
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada
| | - Emma Brasell
- Department of Human Genetics, McGill University, Montreal, Canada
| | | | - Paul R Goodyer
- Research Institute of the McGill University Health Center, 1001 Décarie Boulevard, EM1.2232, Montreal, QC, H4A 3J1, Canada. .,Department of Experimental Medicine, McGill University, Montreal, Canada. .,Department of Human Genetics, McGill University, Montreal, Canada. .,Department of Pediatrics, McGill University, Montreal, Canada.
| |
Collapse
|
19
|
Boyer O, Bérody S. Congenital nephrotic syndrome: is early aggressive treatment needed?-No. Pediatr Nephrol 2020; 35:1991-1996. [PMID: 32462257 DOI: 10.1007/s00467-020-04556-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 11/27/2022]
Abstract
The management of infants with congenital nephrotic syndrome (CNS) is very challenging as they are prone to severe complications such as hemodynamic disturbances, infections, thromboses, and impaired growth, and most will develop end-stage kidney disease (ESKD) within a few years. Since the seventies, an "aggressive" approach, including daily albumin infusions, early nephrectomies, dialysis, and transplantation, has dramatically improved survival and morbidity. More recent case-note reviews have reported successful conservative treatment (using optimized nutrition, complication prophylaxis, and delayed renal replacement therapy), which led to similarly good outcomes and low complication rates. This questions the indications for early preemptive bilateral nephrectomy and dialysis given the mortality and morbidity rates in dialysis in infants and their life-long management with possible repeated transplantations. Two large series provide the most recent evidences supporting the conservative management: firstly, at least 55% children with CNS are not spontaneously in ESKD at the age of 2 years; secondly, albumin tapering/discontinuation and hospital discharge are possible before nephrectomy; and lastly, CNS complication rates are similar in case of preemptive nephrectomies or conservative care. Until now, no clear genotype-phenotype correlation has been identified to guide clinical management. Taken together, these data support the safety of conservative care until ESKD in a subset of patients with CNS.
Collapse
Affiliation(s)
- Olivia Boyer
- Néphrologie Pédiatrique, Centre de Référence MARHEA, Centre de Référence du Syndrome Néphrotique Idiopathique de l'enfant et l'adulte, Hôpital Necker - Enfants Malades, APHP, Inserm U1163, Institut Imagine, Université de Paris, Paris, France.
| | - Sandra Bérody
- Unité de Soins Intensifs et Réanimation Néonatale, Centre Hospitalier Sud Francilien, Corbeil-Essonnes, France
| |
Collapse
|
20
|
A descriptive study of NPHS1 and NPHS2 mutations in children with congenital nephrotic syndrome. GENE REPORTS 2020. [DOI: 10.1016/j.genrep.2020.100722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
21
|
Schapiro D, Daga A, Lawson JA, Majmundar AJ, Lovric S, Tan W, Warejko JK, Fessi I, Rao J, Airik M, Gee HY, Schneider R, Widmeier E, Hermle T, Ashraf S, Jobst-Schwan T, van der Ven AT, Nakayama M, Shril S, Braun DA, Hildebrandt F. Panel sequencing distinguishes monogenic forms of nephritis from nephrosis in children. Nephrol Dial Transplant 2019; 34:474-485. [PMID: 30295827 DOI: 10.1093/ndt/gfy050] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/21/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Alport syndrome (AS) and atypical hemolytic-uremic syndrome (aHUS) are rare forms of chronic kidney disease (CKD) that can lead to a severe decline of renal function. Steroid-resistant nephrotic syndrome (SRNS) is more common than AS and aHUS and causes 10% of childhood-onset CKD. In recent years, multiple monogenic causes of AS, aHUS and SRNS have been identified, but their relative prevalence has yet to be studied together in a typical pediatric cohort of children with proteinuria and hematuria. We hypothesized that identification of causative mutations by whole exome sequencing (WES) in known monogenic nephritis and nephrosis genes would allow distinguishing nephritis from nephrosis in a typical pediatric group of patients with both proteinuria and hematuria at any level. METHODS We therefore conducted an exon sequencing (WES) analysis for 11 AS, aHUS and thrombotic thrombocytopenic purpura-causing genes in an international cohort of 371 patients from 362 families presenting with both proteinuria and hematuria before age 25 years. In parallel, we conducted either WES or high-throughput exon sequencing for 23 SRNS-causing genes in all patients. RESULTS We detected pathogenic mutations in 18 of the 34 genes analyzed, leading to a molecular diagnosis in 14.1% of families (51 of 362). Disease-causing mutations were detected in 3 AS-causing genes (4.7%), 3 aHUS-causing genes (1.4%) and 12 NS-causing genes (8.0%). We observed a much higher mutation detection rate for monogenic forms of CKD in consanguineous families (35.7% versus 10.1%). CONCLUSIONS We present the first estimate of relative frequency of inherited AS, aHUS and NS in a typical pediatric cohort with proteinuria and hematuria. Important therapeutic and preventative measures may result from mutational analysis in individuals with proteinuria and hematuria.
Collapse
Affiliation(s)
- David Schapiro
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ankana Daga
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jennifer A Lawson
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amar J Majmundar
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Svjetlana Lovric
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Weizhen Tan
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jillian K Warejko
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Inés Fessi
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jia Rao
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Merlin Airik
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Heon Yung Gee
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ronen Schneider
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Eugen Widmeier
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tobias Hermle
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shazia Ashraf
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Tilman Jobst-Schwan
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amelie T van der Ven
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Makiko Nakayama
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Shirlee Shril
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniela A Braun
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Friedhelm Hildebrandt
- Department of Medicine, Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
22
|
Zununi Vahed S, Moghaddas Sani H, Haghi M, Mohajel Shoja M, Ardalan M. TRPC6 and NPHS2 gene variants in adult patients with steroid-resistant nephrotic syndrome in North-West of Iran. Mol Biol Rep 2019; 46:6339-6344. [PMID: 31529341 DOI: 10.1007/s11033-019-05074-1] [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: 05/08/2019] [Accepted: 09/10/2019] [Indexed: 10/26/2022]
Abstract
Podocyte gene mutations and their role in the development of nephrotic syndrome (NS) have been reported in some ethnic groups. The aim of this study was to evaluate the presence of possible variants in TRCP6 and NPHS2 (podocin) genes and their association with clinical manifestations in a group of adult patients with steroid resistant nephrotic syndrome (SRNS). All participants including 36 patients with SRNS and 71 healthy volunteers were genotyped using polymerase chain reaction (PCR) and direct sequencing. Whole exons of NPHS2 gene and -254 C > G, -218 C > T, and -361 A > T polymorphisms in the promoter of TRPC6 gene were studied. There were no significant differences in the allele and genotype frequencies of aforementioned TRCP6 polymorphisms between cases and controls (P > 0.05). However, four novel polymorphisms including - 257 T > C, - 266 G > A, - 293 G > C, and - 21 G > A found in the promoter region of TRPC6 gene that may be involved in SRNS in our cohort. In NPHS2 gene, three different polymorphisms in the NPHS2 gene were found in 7 patients with FSGS and none of the previously reported risk polymorphisms was detected in our patients. Podocin related mutations are not too much associated with SRNS in adults, but we should consider the possibility of TRPC6 gene mutation in this population.
Collapse
Affiliation(s)
| | - Hakimeh Moghaddas Sani
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, 2006, Australia
| | - Mehdi Haghi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | | |
Collapse
|
23
|
Zhu X, Ye Y, Xu C, Gao C, Zhang Y, Zhou J, Lin W, Mao J. Protein phosphatase 2A modulates podocyte maturation and glomerular functional integrity in mice. Cell Commun Signal 2019; 17:91. [PMID: 31387591 PMCID: PMC6685276 DOI: 10.1186/s12964-019-0402-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 07/22/2019] [Indexed: 01/08/2023] Open
Abstract
Background Protein phosphorylation & dephosphorylation are ubiquitous cellular processes that allow for the nuanced and reversible regulation of protein activity. Protein phosphatase 2A (PP2A) is a multifunction phosphatase that is well expressed in all cell types of kidney during early renal development, though its functions in kidney remains to be elucidated. Methods PP2A conditional knock-out mice was generated with PP2A fl/fl mice that were crossed with Podocin-Cre mice. The phenotype of Pod-PP2A–KO mice (homozygous for the floxed PP2A allele with Podocin-Cre) and littermate PP2A fl/fl controls (homozygous for the PP2A allele but lacking Podocin-Cre) were further studied. Primary podocytes isolated from the Pod-PP2A-KO mice were cultured and they were then employed with sing label-free nano-LC − MS/MS technology on a Q-exactive followed by SIEVE processing to identify possible target molecular entities for the dephosphorylation effect of PP2A, in which Western blot and immunofluorescent staining were used to analyze further. Results Pod-PP2A–KO mice were developed with weight loss, growth retardation, proteinuria, glomerulopathy and foot process effacement, together with reduced expression of some slit diaphragm molecules and cytoskeleton rearrangement of podocytes. Y box protein 1 (YB-1) was identified to be the target molecule for dephosphorylation effect of PP2A. Furthermore, YB-1 phosphorylation was up-regulated in the Pod-PP2A–KO mice in contrast to the wild type controls, while total and un-phosphorylated YB-1 both was moderately down-regulated in podocytes from the Pod-PP2A-KO mice. Conclusion Our study revealed the important role of PP2A in regulating the development of foot processes and fully differentiated podocytes whereas fine-tuning of YB-1 via a post-translational modification by PP2A regulating its activity might be crucial for the functional integrity of podocytes and glomerular filtration barrier. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s12964-019-0402-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiujuan Zhu
- Department of Nephrology, The Children Hospital of Zhejiang University School of Medicine, #57 Zhugan Lane, Hangzhou, 310003, Zhejiang Province, People's Republic of China
| | - Yuhong Ye
- Department of Nephrology, The Children Hospital of Zhejiang University School of Medicine, #57 Zhugan Lane, Hangzhou, 310003, Zhejiang Province, People's Republic of China
| | - Chengxian Xu
- Department of Nephrology, The Children Hospital of Zhejiang University School of Medicine, #57 Zhugan Lane, Hangzhou, 310003, Zhejiang Province, People's Republic of China
| | - Cunji Gao
- Chronic Disease Research Institute, Department of Nutrition and Food Hygiene, Zhejiang University School of Public Health, Hangzhou, 310058, Zhejiang Province, People's Republic of China
| | - Yingying Zhang
- Department of Nephrology, The Children Hospital of Zhejiang University School of Medicine, #57 Zhugan Lane, Hangzhou, 310003, Zhejiang Province, People's Republic of China
| | - Jing Zhou
- Harvard Center for Polycystic Kidney Disease Research and Renal Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA02115, USA
| | - Weiqiang Lin
- Institute of Translational Medicine, School of Medicine, Zhejiang University, Hangzhou, 310058, Zhejiang Province, People's Republic of China.
| | - Jianhua Mao
- Department of Nephrology, The Children Hospital of Zhejiang University School of Medicine, #57 Zhugan Lane, Hangzhou, 310003, Zhejiang Province, People's Republic of China.
| |
Collapse
|
24
|
Gribouval O, Boyer O, Hummel A, Dantal J, Martinez F, Sberro-Soussan R, Etienne I, Chauveau D, Delahousse M, Lionet A, Allard J, Pouteil Noble C, Tête MJ, Heidet L, Antignac C, Servais A. Identification of genetic causes for sporadic steroid-resistant nephrotic syndrome in adults. Kidney Int 2019; 94:1013-1022. [PMID: 30348286 DOI: 10.1016/j.kint.2018.07.024] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 06/18/2018] [Accepted: 07/12/2018] [Indexed: 01/15/2023]
Abstract
Monogenic forms of Steroid-Resistant Nephrotic Syndrome (SRNS) have been widely characterized, but genetic screening paradigms preferentially address congenital, infantile onset, and familial cases. Our aim was to characterize the distribution of disease-causing gene mutations in adults with sporadic SRNS or focal segmental glomerulosclerosis (FSGS). We selected adult patients with non-syndromic, biopsy-proven FSGS or SRNS in the absence of known family history. Strict clinical criteria included lack of response to glucocorticoids and cyclosporine, and no recurrence after kidney transplantation. Mutations in SRNS genes were detected using a targeted gene panel. Sixteen of 135 tested participants (11.8%) carried pathogenic mutations in monogenic SRNS genes, and 14 others (10.4%) carried two APOL1 high-risk alleles. Autosomal recessive disease was diagnosed in 5 participants, autosomal dominant disease in 9, and X-linked disease in 2. Four participants carried a de novo heterozygous mutation. Among the 16 participants with identified mutations in monogenic SNRS genes, 7 (43.7%) had type IV collagen mutations. Mutations in monogenic SNRS genes were identified primarily in participants with proteinuria onset before 25 years of age, while the age at disease onset was variable in those with APOL1 high-risk genotype. Mean age at diagnosis was lower and renal survival was worse in participants with identified mutations in SNRS genes than in those without mutations. We found a significant rate of pathogenic mutations in adults with SRNS, with Type IV collagen mutations being the most frequent. These findings may have immediate impact on clinical practice.
Collapse
Affiliation(s)
- Olivier Gribouval
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Olivia Boyer
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France; Pediatric Nephrology, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Necker Hospital, Paris, France
| | - Aurélie Hummel
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France; Adult Nephrology & Transplantation, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Necker Hospital, Paris, France
| | | | - Frank Martinez
- Adult Nephrology & Transplantation, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Necker Hospital, Paris, France
| | - Rebecca Sberro-Soussan
- Adult Nephrology & Transplantation, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Necker Hospital, Paris, France
| | - Isabelle Etienne
- Nephrology Department, Hôpital de Bois-Guillaume, CHU Rouen, Rouen, France
| | - Dominique Chauveau
- Department of Nephrology and Organ Transplantation, CHU Rangueil, Toulouse, France
| | | | - Arnaud Lionet
- Nephrology Department, Hôpital Huriez, CHU Lille, Lille, France
| | | | - Claire Pouteil Noble
- Nephrology-Transplantation Department, Hôpital Edouard Herriot, Lyon I University, Lyon, France
| | - Marie-Josèphe Tête
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France
| | - Laurence Heidet
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France; Pediatric Nephrology, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Necker Hospital, Paris, France; Genetic Department, Necker Hospital, APHP, Paris, France
| | - Corinne Antignac
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France; Genetic Department, Necker Hospital, APHP, Paris, France.
| | - Aude Servais
- Inserm U1163, Imagine Institute, Paris Descartes University, Paris, France; Adult Nephrology & Transplantation, Centre de référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Necker Hospital, Paris, France.
| |
Collapse
|
25
|
Multifactorial Origin of Exertional Rhabdomyolysis, Recurrent Hematuria, and Episodic Pain in a Service Member with Sickle Cell Trait. Case Rep Genet 2018; 2018:6898546. [PMID: 30533233 PMCID: PMC6247656 DOI: 10.1155/2018/6898546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 10/24/2018] [Indexed: 11/20/2022] Open
Abstract
Individuals with Sickle Cell Trait (SCT), generally considered a benign carrier state of hemoglobin S (HbAS), are thought to be at risk for exertional rhabdomyolysis and hematuria, conditions that can also be caused by various other acquired and inherited factors. We report an SCT positive service member with an exertional rhabdomyolysis event, recurrent hematuria with transient proteinuria, and episodic burning pain in the lower extremities. Clinical and genetic studies revealed the multifactorial nature of his complex phenotype. The service member was taking prescription medications known to be associated with exertional rhabdomyolysis. He carried a pathogenic mutation, NPHS2 p.V260E, reported in nephropathy and a new variant p.R838Q in SCN11A, a gene involved in familial episodic pain syndrome. Results suggest that drug-to-drug interactions coupled with the stress of exercise, coinheritance of HbAS and NPHS2 p.V260E, and p. R838Q in SCN11A contributed to exertional rhabdomyolysis, recurrent hematuria with proteinuria, and episodic pain, respectively. This case underscores the importance of comprehensive clinical and genetic evaluations to identify underlying causes of health complications reported in SCT individuals.
Collapse
|
26
|
Yu SMW, Nissaisorakarn P, Husain I, Jim B. Proteinuric Kidney Diseases: A Podocyte's Slit Diaphragm and Cytoskeleton Approach. Front Med (Lausanne) 2018; 5:221. [PMID: 30255020 PMCID: PMC6141722 DOI: 10.3389/fmed.2018.00221] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/18/2018] [Indexed: 01/19/2023] Open
Abstract
Proteinuric kidney diseases are a group of disorders with diverse pathological mechanisms associated with significant losses of protein in the urine. The glomerular filtration barrier (GFB), comprised of the three important layers, the fenestrated glomerular endothelium, the glomerular basement membrane (GBM), and the podocyte, dictates that disruption of any one of these structures should lead to proteinuric disease. Podocytes, in particular, have long been considered as the final gatekeeper of the GFB. This specialized visceral epithelial cell contains a complex framework of cytoskeletons forming foot processes and mediate important cell signaling to maintain podocyte health. In this review, we will focus on slit diaphragm proteins such as nephrin, podocin, TRPC6/5, as well as cytoskeletal proteins Rho/small GTPases and synaptopodin and their respective roles in participating in the pathogenesis of proteinuric kidney diseases. Furthermore, we will summarize the potential therapeutic options targeting the podocyte to treat this group of kidney diseases.
Collapse
Affiliation(s)
- Samuel Mon-Wei Yu
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States
| | | | - Irma Husain
- Department of Medicine, James J. Peters VA Medical Center, Bronx, NY, United States
| | - Belinda Jim
- Department of Medicine, Jacobi Medical Center, Bronx, NY, United States.,Renal Division, Jacobi Medical Center, Bronx, NY, United States
| |
Collapse
|
27
|
Asharam K, Bhimma R, David VA, Coovadia HM, Qulu WP, Naicker T, Gillies CE, Vega-Warner V, Johnson RC, Limou S, Kopp JB, Sampson M, Nelson GW, Winkler CA. NPHS2 V260E Is a Frequent Cause of Steroid-Resistant Nephrotic Syndrome in Black South African Children. Kidney Int Rep 2018; 3:1354-1362. [PMID: 30450462 PMCID: PMC6224675 DOI: 10.1016/j.ekir.2018.07.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Revised: 07/12/2018] [Accepted: 07/23/2018] [Indexed: 11/23/2022] Open
Abstract
Introduction In South Africa (SA), steroid-resistant nephrotic syndrome (SRNS) is more frequent in black than in Indian children. Methods Seeking a genetic basis for this disparity, we enrolled 33 Indian and 31 black children with steroid-sensitive nephrotic syndrome (SSNS) and SRNS from KwaZulu-Natal, SA; SRNS children underwent kidney biopsy. We sequenced NPHS2 and genotyped APOL1 in 15 SSNS and 64 SRNS unrelated patients and 104 controls and replicated results in 18 black patients with steroid-resistant focal segmental glomerulosclerosis (SR-FSGS). Known FSGS genes (n = 21) were sequenced in a subset of patients. Results Homozygosity for NPHS2 V260E was found in 8 of 30 black children with SRNS (27%); all 260E/E carriers had SR-FSGS. Combining SR-FSGS patients from the 2 groups, 14 of 42 (33%) were homozygous for V260E. One black control was heterozygous for V260E; no Indian patients or controls were carriers. Haplotype analysis indicated that homozygosity for V260E was not explained by cryptic consanguinity. Children with NPHS2 260E/E developed SRNS at earlier age than noncarriers (34 vs. 78 months, P = 0.01), and none achieved partial or complete remission (0% vs. 47%, P = 0.002). APOL1 variants did not associate with NS. Sequencing FSGS genes identified a CD2AP predicted pathogenic variant in the heterozygous state in 1 Indian case with SR-FSGS. Conclusion NPHS2 260E/E was present in one-third of black FSGS patients, was absent in black controls and Indian patients, and affected patients were unresponsive to therapy. Genotyping V260E in black children from South Africa with NS will identify a substantial group with SR-FSGS, potentially sparing these children biopsy and ineffective steroid treatment.
Collapse
Affiliation(s)
| | | | - Victor A. David
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, Maryland, USA
| | - Hoosen M. Coovadia
- University of KwaZulu-Natal, Durban, South Africa
- School of Public Health, University of Witwatersrand, Johannesburg, South Africa
| | | | | | - Christopher E. Gillies
- Department of Pediatrics-Nephrology; University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Virginia Vega-Warner
- Department of Pediatrics-Nephrology; University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Randall C. Johnson
- Advanced Biomedical Computing Center, Frederick National Laboratory, Frederick, Maryland, USA
| | - Sophie Limou
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland, USA
| | - Mathew Sampson
- Department of Pediatrics-Nephrology; University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - George W. Nelson
- Advanced Biomedical Computing Center, Frederick National Laboratory, Frederick, Maryland, USA
| | - Cheryl A. Winkler
- Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Leidos Biomedical Research, Inc., Frederick National Laboratory, Frederick, Maryland, USA
- Correspondence: Cheryl A. Winkler, 8560 Progress Drive, National Cancer Institute, Frederick National Laboratory, Frederick, Maryland 21701, USA.
| |
Collapse
|
28
|
Estrella MM, Parekh RS. The Expanding Role of APOL1 Risk in Chronic Kidney Disease and Cardiovascular Disease. Semin Nephrol 2018; 37:520-529. [PMID: 29110759 DOI: 10.1016/j.semnephrol.2017.07.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Variants of the APOL1 gene, found primarily in individuals of African descent, are associated with various forms of kidney disease and kidney disease progression. Recent studies evaluating the association of APOL1 with cardiovascular disease have yielded conflicting results, and the potential role in cardiovascular disease remains unclear. In this review, we summarize the observational studies linking the APOL1 risk variants with chronic kidney and cardiovascular disease among persons of African descent.
Collapse
Affiliation(s)
- Michelle M Estrella
- Kidney Health Research Collaborative, Department of Medicine, University of California San Francisco, San Francisco, CA; Department of Medicine, San Francisco VA Medical Center, San Francisco, CA
| | - Rulan S Parekh
- Division of Nephrology, Departments of Pediatrics and Medicine, The Hospital for Sick Children, SickKids Research Institute, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
29
|
Stráner P, Balogh E, Schay G, Arrondel C, Mikó Á, L'Auné G, Benmerah A, Perczel A, K Menyhárd D, Antignac C, Mollet G, Tory K. C-terminal oligomerization of podocin mediates interallelic interactions. Biochim Biophys Acta Mol Basis Dis 2018; 1864:2448-2457. [PMID: 29660491 DOI: 10.1016/j.bbadis.2018.04.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Revised: 03/22/2018] [Accepted: 04/11/2018] [Indexed: 01/01/2023]
Abstract
Interallelic interactions of membrane proteins are not taken into account while evaluating the pathogenicity of sequence variants in autosomal recessive disorders. Podocin, a membrane-anchored component of the slit diaphragm, is encoded by NPHS2, the major gene mutated in hereditary podocytopathies. We formerly showed that its R229Q variant is only pathogenic when trans-associated to specific 3' mutations and suggested the causal role of an abnormal C-terminal dimerization. Here we show by FRET analysis and size exclusion chromatography that podocin oligomerization occurs exclusively through the C-terminal tail (residues 283-382): principally through the first C-terminal helical region (H1, 283-313), which forms a coiled coil as shown by circular dichroism spectroscopy, and through the 332-348 region. We show the principal role of the oligomerization sites in mediating interallelic interactions: while the monomer-forming R286Tfs*17 podocin remains membranous irrespective of the coexpressed podocin variant identity, podocin variants with an intact H1 significantly influence each other's localization (r2 = 0.68, P = 9.2 × 10-32). The dominant negative effect resulting in intracellular retention of the pathogenic F344Lfs*4-R229Q heterooligomer occurs in parallel with a reduction in the FRET efficiency, suggesting the causal role of a conformational rearrangement. On the other hand, oligomerization can also promote the membrane localization: it can prevent the endocytosis of F344Lfs*4 or F344* podocin mutants induced by C-terminal truncation. In conclusion, C-terminal oligomerization of podocin can mediate both a dominant negative effect and interallelic complementation. Interallelic interactions of NPHS2 are not restricted to the R229Q variant and have to be considered in compound heterozygous individuals.
Collapse
Affiliation(s)
- Pál Stráner
- MTA-ELTE Protein Modeling Research Group and Laboratory of Structural Chemistry and Biology, Eötvös Loránd University, Budapest, Hungary
| | - Eszter Balogh
- MTA-SE Lendület Nephrogenetic Laboratory, Budapest, Hungary; Semmelweis University, Ist Department of Pediatrics, Budapest, Hungary
| | - Gusztáv Schay
- Semmelweis University, Department of Biophysics and Radiation Biology, Budapest, Hungary
| | - Christelle Arrondel
- Laboratory of Hereditary Kidney Diseases, INSERM, UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Ágnes Mikó
- MTA-SE Lendület Nephrogenetic Laboratory, Budapest, Hungary; Semmelweis University, Ist Department of Pediatrics, Budapest, Hungary
| | - Gerda L'Auné
- MTA-SE Lendület Nephrogenetic Laboratory, Budapest, Hungary; Semmelweis University, Ist Department of Pediatrics, Budapest, Hungary
| | - Alexandre Benmerah
- Laboratory of Hereditary Kidney Diseases, INSERM, UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - András Perczel
- MTA-ELTE Protein Modeling Research Group and Laboratory of Structural Chemistry and Biology, Eötvös Loránd University, Budapest, Hungary
| | - Dóra K Menyhárd
- MTA-ELTE Protein Modeling Research Group and Laboratory of Structural Chemistry and Biology, Eötvös Loránd University, Budapest, Hungary
| | - Corinne Antignac
- Laboratory of Hereditary Kidney Diseases, INSERM, UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France; Assistance Publique - Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Département de Génétique, Paris, France
| | - Géraldine Mollet
- Laboratory of Hereditary Kidney Diseases, INSERM, UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Kálmán Tory
- MTA-SE Lendület Nephrogenetic Laboratory, Budapest, Hungary; Semmelweis University, Ist Department of Pediatrics, Budapest, Hungary; Laboratory of Hereditary Kidney Diseases, INSERM, UMR 1163, Imagine Institute, Paris, France; Université Paris Descartes-Sorbonne Paris Cité, Imagine Institute, Paris, France.
| |
Collapse
|
30
|
Lepori N, Zand L, Sethi S, Fernandez-Juarez G, Fervenza FC. Clinical and pathological phenotype of genetic causes of focal segmental glomerulosclerosis in adults. Clin Kidney J 2018; 11:179-190. [PMID: 29644057 PMCID: PMC5888331 DOI: 10.1093/ckj/sfx143] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023] Open
Abstract
Focal segmental glomerulosclerosis (FSGS) is a histologic lesion resulting from a variety of pathogenic processes that cause injury to the podocytes. Recently, mutations in more than 50 genes expressed in podocyte or glomerular basement membrane were identified as causing genetic forms of FSGS, the majority of which are characterized by onset in childhood. The prevalence of adult-onset genetic FSGS is likely to be underestimated and its clinical and histological features have not been clearly described. A small number of studies of adult-onset genetic FSGS showed that there is heterogeneity in clinical and histological findings, with a presentation ranging from sub-nephrotic proteinuria to full nephrotic syndrome. A careful evaluation of adult-onset FSGS that do not have typical features of primary or secondary FSGS (familial cases, resistance to immunosuppression and absence of evident cause of secondary FSGS) should include a genetic evaluation. Indeed, recognizing genetic forms of adult-onset FSGS is of the utmost importance, given that this diagnosis will have major implications on treatment strategies, selecting of living-related kidney donor and renal transplantation success.
Collapse
Affiliation(s)
- Nicola Lepori
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Division of Nephrology and Dialysis, Azienda Ospedaliera G. Brotzu, Cagliari, Italy
| | - Ladan Zand
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Sanjeev Sethi
- Division of Anatomic Pathology, Mayo Clinic, Rochester, MN, USA
| | - Gema Fernandez-Juarez
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
- Nephrology Division, Hospital Universitario Fundacion Alcorcon, Madrid, Spain
| | | |
Collapse
|
31
|
Martin CE, Jones N. Nephrin Signaling in the Podocyte: An Updated View of Signal Regulation at the Slit Diaphragm and Beyond. Front Endocrinol (Lausanne) 2018; 9:302. [PMID: 29922234 PMCID: PMC5996060 DOI: 10.3389/fendo.2018.00302] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Podocytes are a major component of the glomerular blood filtration barrier, and alterations to the morphology of their unique actin-based foot processes (FP) are a common feature of kidney disease. Adjacent FP are connected by a specialized intercellular junction known as the slit diaphragm (SD), which serves as the ultimate barrier to regulate passage of macromolecules from the blood. While the link between SD dysfunction and reduced filtration selectivity has been recognized for nearly 50 years, our understanding of the underlying molecular circuitry began only 20 years ago, sparked by the identification of NPHS1, encoding the transmembrane protein nephrin. Nephrin not only functions as the core component of the extracellular SD filtration network but also as a signaling scaffold via interactions at its short intracellular region. Phospho-regulation of several conserved tyrosine residues in this region influences signal transduction pathways which control podocyte cell adhesion, shape, and survival, and emerging studies highlight roles for nephrin phospho-dynamics in mechanotransduction and endocytosis. The following review aims to summarize the last 5 years of advancement in our knowledge of how signaling centered at nephrin directs SD barrier formation and function. We further provide insight on promising frontiers in podocyte biology, which have implications for SD signaling in the healthy and diseased kidney.
Collapse
|
32
|
Feltran LS, Varela P, Silva ED, Veronez CL, Franco MC, Filho AP, Camargo MF, Koch Nogueira PC, Pesquero JB. Targeted Next-Generation Sequencing in Brazilian Children With Nephrotic Syndrome Submitted to Renal Transplant. Transplantation 2017; 101:2905-2912. [PMID: 28658201 DOI: 10.1097/tp.0000000000001846] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND The aims of this study were to identify the genetic mutations profile in Brazilian children with nephrotic syndrome (NS) and to determine a genotype-phenotype correlation in this disease. METHODS Next-generation sequencing and mutation analysis were performed on 24 genes related to NS in a cross-sectional study involving 95 children who underwent kidney transplantation due to NS, excluding congenital cases. RESULTS A total of 149 variants were identified in 22 of 24 sequenced genes. The mutations were classified as pathogenic, likely pathogenic, likely benign and benign per the chance of causing the disease. NPHS2 was the most common mutated gene. We identified 8 (8.4%) patients with hereditary NS and 5 (5%) patients with probably genetically caused NS. COL4A3-5 variants were found as well, but it is not clear whether they should be considered isolated FSGS or simply a misdiagnosed type of the Alport spectrum. Considering the clinical results, hereditary NS patients presented a tendency to early disease onset when compared with the other groups (P = 0.06) and time to end stage renal disease (ESRD) was longer in this group (P = 0.03). No patients from hereditary NS group had NS recurrence after transplantation. CONCLUSIONS This is the first study in children with steroid-resistant NS who underwent kidney transplantation using next-generation sequencing. Considering our results, we believe this study has shed some light to the uncertainties of genotype-phenotype correlation in NS, where several genes cooperate to produce or even to modify the course of the disease.
Collapse
Affiliation(s)
- Luciana S Feltran
- Nephrology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Patricia Varela
- Biophysics Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Elton Dias Silva
- Biophysics Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Camila Lopes Veronez
- Biophysics Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Maria Carmo Franco
- Nephrology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Alvaro Pacheco Filho
- Nephrology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Maria Fernanda Camargo
- Nephrology Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Paulo Cesar Koch Nogueira
- Pediatrics Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Joao Bosco Pesquero
- Biophysics Department, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
33
|
Lemoine S, Cochat P, Bertholet-Thomas A, Levi C, Bonnefoy C, Sellier-Leclerc AL, Bacchetta J. Néphrologie pédiatrique : que doit savoir un néphrologue d’adulte sur ces pathologies ? Nephrol Ther 2017; 13:495-504. [DOI: 10.1016/j.nephro.2017.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 11/25/2022]
|
34
|
Zhang H, Wang F, Liu X, Zhong X, Yao Y, Xiao H. Steroid-resistant nephrotic syndrome caused by co-inheritance of mutations at NPHS1 and ADCK4 genes in two Chinese siblings. Intractable Rare Dis Res 2017; 6:299-303. [PMID: 29259860 PMCID: PMC5735285 DOI: 10.5582/irdr.2017.01037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Hereditary nephrotic syndrome often presents with steroid-resistance and onset within the first year of life. Mutations in genes highly expressed in podocytes have been found in two thirds of these patients, especially NPHS1 and NPHS2 among at least 29 genetic causes that have been discovered. We reported two siblings with steroid-resistant nephrotic syndrome caused by co-inheritance of mutations at NPHS1 (c.1339G>A, p.E447K) and ACDK4 (c.748G>C, p.D250H) genes. The siblings presented with steroid-resistant nephrotic syndrome and pathological lesions of focal segmental glomerulosclerosis (FSGS), while the elder sister also developed hypertension, renal failure and cardiac dysfunction.
Collapse
Affiliation(s)
- Hongwen Zhang
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Fang Wang
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Xiaoyu Liu
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Xuhui Zhong
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Yong Yao
- Department of Pediatric, Peking University First Hospital, Beijing, China
| | - Huijie Xiao
- Department of Pediatric, Peking University First Hospital, Beijing, China
- Address correspondence to: Dr. Huijie Xiao, Department of Pediatric, Peking University First Hospital, No.1 Xi An Men Da Jie, Beijing 100034, China. E-mail:
| |
Collapse
|
35
|
Tabatabaeifar M, Wlodkowski T, Simic I, Denc H, Mollet G, Weber S, Moyers JJ, Brühl B, Randles MJ, Lennon R, Antignac C, Schaefer F. An inducible mouse model of podocin-mutation-related nephrotic syndrome. PLoS One 2017; 12:e0186574. [PMID: 29049388 PMCID: PMC5648285 DOI: 10.1371/journal.pone.0186574] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 09/10/2017] [Indexed: 12/03/2022] Open
Abstract
Mutations in the NPHS2 gene, encoding podocin, cause hereditary nephrotic syndrome. The most common podocin mutation, R138Q, is associated with early disease onset and rapid progression to end-stage renal disease. Knock-in mice carrying a R140Q mutation, the mouse analogue of human R138Q, show developmental arrest of podocytes and lethal renal failure at neonatal age. Here we created a conditional podocin knock-in model named NPHS2R140Q/-, using a tamoxifen-inducible Cre recombinase, which permits to study the effects of the mutation in postnatal life. Within the first week of R140Q hemizygosity induction the animals developed proteinuria, which peaked after 4–5 weeks. Subsequently the animals developed progressive renal failure, with a median survival time of 12 (95% CI: 11–13) weeks. Foot process fusion was observed within one week, progressing to severe and global effacement in the course of the disease. The number of podocytes per glomerulus gradually diminished to 18% compared to healthy controls 12–16 weeks after induction. The fraction of segmentally sclerosed glomeruli was 25%, 85% and 97% at 2, 4 and 8 weeks, respectively. Severe tubulointerstitial fibrosis was present at later disease stage and was correlated quantitatively with the level of proteinuria at early disease stages. While R140Q podocin mRNA expression was elevated, protein abundance was reduced by more than 50% within one week following induction. Whereas miRNA21 expression persistently increased during the first 4 weeks, miRNA-193a expression peaked 2 weeks after induction. In conclusion, the inducible R140Q-podocin mouse model is an auspicious model of the most common genetic cause of human nephrotic syndrome, with a spontaneous disease course strongly reminiscent of the human disorder. This model constitutes a valuable tool to test the efficacy of novel pharmacological interventions aimed to improve podocyte function and viability and attenuate proteinuria, glomerulosclerosis and progressive renal failure.
Collapse
Affiliation(s)
- Mansoureh Tabatabaeifar
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Tanja Wlodkowski
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Ivana Simic
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Helga Denc
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
| | - Geraldine Mollet
- INSERM, U1163, Imagine Institute, Laboratory of Hereditary Kidney Diseases, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Paris, France
| | - Stefanie Weber
- Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Philipps-University Marburg, Marburg, Germany
| | | | - Barbara Brühl
- Institute for Anatomy and Cell Biology, University of Heidelberg, Heidelberg, Germany
| | - Michael Joseph Randles
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Rachel Lennon
- Wellcome Trust Centre for Cell Matrix Research, University of Manchester, Manchester, United Kingdom
- Faculty of Biology Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Corinne Antignac
- INSERM, U1163, Imagine Institute, Laboratory of Hereditary Kidney Diseases, Paris, France
- Paris Descartes-Sorbonne Paris Cité University, Paris, France
- Department of Genetics, Necker Hospital, Assistance Publique—Hôpitaux de Paris, Paris, France
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| |
Collapse
|
36
|
Joshi BB, Mistry KN, Gang S, Koringa PG, Joshi CG. Characterization of NPHS2 gene polymorphisms associated to steroid resistance nephrotic syndrome in Indian children. Gene 2017; 628:134-140. [PMID: 28712774 DOI: 10.1016/j.gene.2017.07.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 06/02/2017] [Accepted: 07/10/2017] [Indexed: 11/18/2022]
Abstract
Nephrotic syndrome (NS) is the common glomerular disease in children. These children are treated with steroids, depending upon their behavior. They are either steroid sensitive (SSNS) or steroid resistant (SRNS). NPHS2 gene mutants are linked to the risk of autosomal recessive SRNS and in some cases to SSNS. The present study has been performed to screen single nucleotide polymorphisms (SNPs) of the NPHS2 gene in a group of 90 Indian children suffering with NS (30 SSNS, 30 SRNS and 30 Controls) by PCR method followed by direct exon sequencing. Effect of SNPs on fold expression changes at transcript level of podocin was checked using quantitative real time PCR (qRT-PCR). SNPs identified through sequencing helps to carry out in-silico analysis. Overall 17 SNPs were identified in NPHS2 gene where 6 were found novel. Three missense SNPs p.R299Q, p.P20L and p.G35D were also identified in this population where SNP, p.G35D was found novel. In addition to sequencing analysis, results of in silico analysis shows that a mutant with these three missense SNPs has least ligand binding efficiency compared to native model. Moreover the significant observation of this study included two intronic SNPs c.451+23C>T and c.451+58A>T present in SRNS group of patients. These SNPs has shown high level of clinical significance within genomic and allelic frequency along with haplotypes and linkage disequilibrium count. The qRT-PCR analysis shows, down expression of podocin protein at transcript level in SRNS patients compared to SSNS patients. All these results support the fact that SNPs present in this population could affect the protein structural stability. Thus it is concluded that the polymorphisms predicted in this study might be disease causing in the NPHS2 gene and may have influence on the therapeutic response of NS patients.
Collapse
Affiliation(s)
- Bhoomi B Joshi
- Ashok and Rita Patel Institute of Integrated Study and Research in Biotechnology and Allied Sciences (ARIBAS), Sardar Patel University, Vallabh Vidyanagar, Gujarat 388120, India
| | - Kinnari N Mistry
- Ashok and Rita Patel Institute of Integrated Study and Research in Biotechnology and Allied Sciences (ARIBAS), Sardar Patel University, Vallabh Vidyanagar, Gujarat 388120, India.
| | - Sishir Gang
- Muljibhai Patel Urological Hospital, Dr. V.V. Desai Road, Nadiad 387 001, Gujarat, India
| | - Prakash G Koringa
- College of Veterinary Sciences and Animal Husbandry, Anand Agricultural University, India
| | - Chaitanya G Joshi
- College of Veterinary Sciences and Animal Husbandry, Anand Agricultural University, India
| |
Collapse
|
37
|
NPHS2 Mutations: A Closer Look to Latin American Countries. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7518789. [PMID: 28785586 PMCID: PMC5529630 DOI: 10.1155/2017/7518789] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Revised: 06/02/2017] [Accepted: 06/08/2017] [Indexed: 01/10/2023]
Abstract
Nephrotic syndrome is one of the most common kidney pathologies in childhood, being characterized by proteinuria, edema, and hypoalbuminemia. In clinical practice, it is divided into two categories based on the response to steroid therapy: steroid-sensitive and steroid resistant. Inherited impairments of proteins located in the glomerular filtration barrier have been identified as important causes of nephrotic syndrome, with one of these being podocin, coded by NPHS2 gene. NPHS2 mutations are the most frequent genetic cause of steroid resistant nephrotic syndrome. The aim of this review is to update the list of NPHS2 mutations reported between June 2013 and February 2017, with a closer look to mutations occurring in Latin American countries.
Collapse
|
38
|
Bierzynska A, McCarthy HJ, Soderquest K, Sen ES, Colby E, Ding WY, Nabhan MM, Kerecuk L, Hegde S, Hughes D, Marks S, Feather S, Jones C, Webb NJA, Ognjanovic M, Christian M, Gilbert RD, Sinha MD, Lord GM, Simpson M, Koziell AB, Welsh GI, Saleem MA. Genomic and clinical profiling of a national nephrotic syndrome cohort advocates a precision medicine approach to disease management. Kidney Int 2017; 91:937-947. [PMID: 28117080 DOI: 10.1016/j.kint.2016.10.013] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 09/08/2016] [Accepted: 10/06/2016] [Indexed: 11/30/2022]
Abstract
Steroid Resistant Nephrotic Syndrome (SRNS) in children and young adults has differing etiologies with monogenic disease accounting for 2.9-30% in selected series. Using whole exome sequencing we sought to stratify a national population of children with SRNS into monogenic and non-monogenic forms, and further define those groups by detailed phenotypic analysis. Pediatric patients with SRNS were identified via a national United Kingdom Renal Registry. Whole exome sequencing was performed on 187 patients, of which 12% have a positive family history with a focus on the 53 genes currently known to be associated with nephrotic syndrome. Genetic findings were correlated with individual case disease characteristics. Disease causing variants were detected in 26.2% of patients. Most often this occurred in the three most common SRNS-associated genes: NPHS1, NPHS2, and WT1 but also in 14 other genes. The genotype did not always correlate with expected phenotype since mutations in OCRL, COL4A3, and DGKE associated with specific syndromes were detected in patients with isolated renal disease. Analysis by primary/presumed compared with secondary steroid resistance found 30.8% monogenic disease in primary compared with none in secondary SRNS permitting further mechanistic stratification. Genetic SRNS progressed faster to end stage renal failure, with no documented disease recurrence post-transplantation within this cohort. Primary steroid resistance in which no gene mutation was identified had a 47.8% risk of recurrence. In this unbiased pediatric population, whole exome sequencing allowed screening of all current candidate genes. Thus, deep phenotyping combined with whole exome sequencing is an effective tool for early identification of SRNS etiology, yielding an evidence-based algorithm for clinical management.
Collapse
Affiliation(s)
- Agnieszka Bierzynska
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK
| | - Hugh J McCarthy
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK
| | - Katrina Soderquest
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Ethan S Sen
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK
| | - Elizabeth Colby
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK
| | - Wen Y Ding
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK
| | - Marwa M Nabhan
- Egyptian group for orphan renal diseases (EGORD), Department of paediatrics, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt
| | | | | | | | | | | | | | - Nicholas J A Webb
- Department of Paediatric Nephrology and NIHR/Wellcome Trust Clinical Research Facility, University of Manchester, Manchester Academic Health Science Centre, Royal Manchester Children's Hospital, Manchester, UK
| | - Milos Ognjanovic
- Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle-upon-Tyne, UK
| | | | - Rodney D Gilbert
- Southampton Children's Hospital and University of Southampton School of Medicine, Southampton, UK
| | | | - Graham M Lord
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Michael Simpson
- Division of Genetics and Molecular Medicine, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Ania B Koziell
- Division of Transplantation Immunology and Mucosal Biology, Department of Experimental Immunobiology, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Gavin I Welsh
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK
| | - Moin A Saleem
- Bristol Renal and Children's Renal Unit, School of Clinical Sciences, University of Bristol, UK.
| |
Collapse
|
39
|
Novel NPHS2 variant in patients with familial steroid-resistant nephrotic syndrome with early onset, slow progression and dominant inheritance pattern. Clin Exp Nephrol 2016; 21:677-684. [DOI: 10.1007/s10157-016-1331-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 08/22/2016] [Indexed: 12/01/2022]
|
40
|
Davin JC. The glomerular permeability factors in idiopathic nephrotic syndrome. Pediatr Nephrol 2016; 31:207-15. [PMID: 25925039 PMCID: PMC4689751 DOI: 10.1007/s00467-015-3082-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Revised: 02/25/2015] [Accepted: 02/27/2015] [Indexed: 12/19/2022]
Abstract
It is currently postulated that steroid-sensitive idiopathic nephrotic syndrome (SSNS) and steroid-resistant idiopathic nephrotic syndrome (SRNS), which are not related to the mutation of a gene coding for podocyte structures or for glomerular basement membrane proteins, result from a circulating factor affecting podocyte shape and function. T lymphocytes have for a long time been suspected to be involved in the pathophysiology of these diseases. The successful treatment of steroid-dependant nephrotic syndrome with rituximab suggests a potential role for B lymphocytes. Clinical and experimental data indicate roles for cytokines IL-13, TNFα, circulating cardiotrophin-like cytokine factor 1 (member of the IL-6 family), circulating hemopexin, radical oxygen species, and the soluble urokinase-type plasminogen activator receptor (suPAR) in the development of nephrotic syndrome. Podocyte metabolism modifications-leading to the overexpression of the podocyte B7-1antigen (CD 80), hypoactivity of the podocyte enzyme sphingomyelin phosphodiesterase acid-like 3 b (SMPDL3b), and to the podocyte production of a hyposialylated form of the angiopoietin-like 4 (Angptl4)-are mechanisms possibly involved in the changes in the podocyte cytoskeleton leading to SSNS and or SRNS. Different multifactorial pathophysiological mechanisms can be advocated for SSNS and SRNS. The present paper reviews the experimental and clinical data upon which the different hypotheses are based and reports their possible clinical applications.
Collapse
Affiliation(s)
- Jean-Claude Davin
- Emma Children's Hospital/ Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands.
- Queen Fabiola Academic Children's Hospital, Free University of Brussels, Brussels, Belgium.
- Pediatric Nephrology Department, Emma Children's Hospital/ Academic Medical Centre, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
41
|
Ranganathan S. Pathology of Podocytopathies Causing Nephrotic Syndrome in Children. Front Pediatr 2016; 4:32. [PMID: 27066465 PMCID: PMC4814732 DOI: 10.3389/fped.2016.00032] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 03/21/2016] [Indexed: 12/31/2022] Open
Abstract
Nephrotic syndrome (NS) in children includes a diverse group of diseases that range from genetic diseases without any immunological defects to causes that are primarily due to immunological effects. Recent advances in molecular and genomic studies have resulted in a plethora of genetic defects that have been localized to the podocyte, the basic structure that is instrumental in normal filtration process. Although the disease can manifest from birth and into adulthood, the primary focus of this review would be to describe the novel genes and pathology of primary podocyte defects that cause NS in children. This review will restrict itself to the pathology of congenital NS, minimal change disease (MCD), and its variants and focal segmental glomerulosclerosis (FSGS). The two major types of congenital NS are Finnish type characterized by dilated sausage shaped tubules morphologically and diffuse mesangial sclerosis characterized by glomerulosclerosis. MCD has usually normal appearing biopsy features on light microscopy and needs electron microscopy for diagnosis, whereas FSGS in contrast has classic segmental sclerosing lesions identified in different portions of the glomeruli and tubular atrophy. This review summarizes the pathological characteristics of these conditions and also delves into the various genetic defects that have been described as the cause of these primary podocytopathies. Other secondary causes of NS in children, such as membranoproliferative and membranous glomerulonephritis, will not be covered in this review.
Collapse
|
42
|
Beltcheva O, Boueva A, Tzveova R, Roussinov D, Marinova S, Kaneva R, Mitev V. Steroid-resistant nephrotic syndrome caused by novel WT1 mutation inherited from a mosaic parent. Ren Fail 2015; 38:290-3. [PMID: 26627896 DOI: 10.3109/0886022x.2015.1117906] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a severe childhood disorder frequently progressing toward renal failure. Among its genetic causes are mutations in the Wilms tumor gene, WT1, which codes for a transcription factor with key role for the embryonic development of the genitourinary tract as well as for maintaining podocyte differentiation and slit diaphragm structure in adults. Defects in WT1 are associated with sporadic cases of both syndromic and isolated SRNS. We report here a novel WT1 mutation associated with SRNS in a female patient, which leads to a Cys428Ser substitution on protein level, affecting one of the cysteine residues responsible for zinc binding in the second zinc finger domain. Surprisingly, the mutation identified in the patient was found to be inherited from the healthy mosaic mother. The presence of mosaicism was confirmed using quantitative polymerase chain reaction (PCR) high-resolution melting. The clinical implications of this finding for the family are discussed.
Collapse
Affiliation(s)
- Olga Beltcheva
- a Molecular Medicine Center , Department of Medical Chemistry and Biochemistry, Medical University of Sofia , Sofia , Bulgaria
| | - Anelia Boueva
- b SBAL Pediatric Diseases, Pediatric Nephrology and Hemodialysis Clinic, Medical University of Sofia , Sofia , Bulgaria
| | - Reni Tzveova
- a Molecular Medicine Center , Department of Medical Chemistry and Biochemistry, Medical University of Sofia , Sofia , Bulgaria
| | - Dimitar Roussinov
- b SBAL Pediatric Diseases, Pediatric Nephrology and Hemodialysis Clinic, Medical University of Sofia , Sofia , Bulgaria
| | - Svetlana Marinova
- b SBAL Pediatric Diseases, Pediatric Nephrology and Hemodialysis Clinic, Medical University of Sofia , Sofia , Bulgaria
| | - Radka Kaneva
- a Molecular Medicine Center , Department of Medical Chemistry and Biochemistry, Medical University of Sofia , Sofia , Bulgaria
| | - Vanio Mitev
- a Molecular Medicine Center , Department of Medical Chemistry and Biochemistry, Medical University of Sofia , Sofia , Bulgaria
| |
Collapse
|
43
|
Guaragna MS, Lutaif ACGB, Piveta CSC, Souza ML, de Souza SR, Henriques TB, Maciel-Guerra AT, Belangero VMS, Guerra-Junior G, De Mello MP. NPHS2 mutations account for only 15% of nephrotic syndrome cases. BMC MEDICAL GENETICS 2015; 16:88. [PMID: 26420286 PMCID: PMC4589073 DOI: 10.1186/s12881-015-0231-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 09/15/2015] [Indexed: 01/27/2023]
Abstract
Background Nephrotic syndrome is traditionally classified on the basis of the response to standard steroid treatment. Mutations in more than 24 genes have been associated with nephrotic syndrome in children, although the great majority of steroid-resistant cases have been attributed to mutations in three main genes: NPHS1, NPHS2 and WT1. The aims of this study were to identify mutations in these genes more frequently reported as mutated and to characterize each variation using different in silico prediction algorithms in order to understand their biological functions. Methods We performed direct sequence analysis of exons 8 and 9 of WT1, 8 exons of NPHS2 and 29 exons of NPHS1, including NPHS2 and NPHS1 intron–exon boundary sequences, as well as 700 bp of the 5′ UTR from both genes in 27 steroid-resistant patients aged between 3 months and 18 years. Results Analysis of the NPHS2 gene revealed four missense mutations, one frameshift mutation and three variations in the 5′ UTR. Four patients presented compound heterozygosis, and four other patients presented one heterozygous alteration only. WT1 and NPHS1 gene analysis did not reveal any mutations. Discussion This is the first study focusing on genetics of SRNS in Brazilian children. Identification of mutations is important because it could influence physicians’ decision on patient treatment, as patients carrying mutations can be spared the side effects of immunosuppressive therapy and ultimately could be considered for kidney transplantation from a living donor. Conclusions After molecular analysis of the genes more frequently reported as mutated in 27 steroid-resistant nephrotic syndrome patients, we identified NPHS2 mutations confirming the hereditary character of the kidney disease in only 14.8 % of patients. Therefore, the next step is to perform a next generation sequencing based analysis of glomeluropathy-related panel of genes for the remaining patients in order to search for mutations in other genes related to steroid-resistant nephrotic syndrome. Electronic supplementary material The online version of this article (doi:10.1186/s12881-015-0231-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Mara Sanches Guaragna
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, São Paulo, Caixa Postal 6010, Brasil.
| | - Anna Cristina G B Lutaif
- Nefrologia Pediátrica, Departamento de Pediatria, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Cristiane S C Piveta
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, São Paulo, Caixa Postal 6010, Brasil. .,Centro de Investigação em Pediatria, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Marcela L Souza
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, São Paulo, Caixa Postal 6010, Brasil.
| | - Suéllen R de Souza
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, São Paulo, Caixa Postal 6010, Brasil.
| | - Taciane B Henriques
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, São Paulo, Caixa Postal 6010, Brasil.
| | - Andréa T Maciel-Guerra
- Departamento de Genética Médica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil. .,Grupo Interdisciplinar de Estudos da Determinação e Diferenciação do Sexo, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Vera M S Belangero
- Nefrologia Pediátrica, Departamento de Pediatria, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Gil Guerra-Junior
- Centro de Investigação em Pediatria, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil. .,Grupo Interdisciplinar de Estudos da Determinação e Diferenciação do Sexo, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil. .,Endocrinologia Pediátrica, Departamento de Pediatria, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brasil.
| | - Maricilda P De Mello
- Centro de Biologia Molecular e Engenharia Genética, Universidade Estadual de Campinas, Campinas, São Paulo, Caixa Postal 6010, Brasil.
| |
Collapse
|
44
|
Rheault MN, Gbadegesin RA. The Genetics of Nephrotic Syndrome. J Pediatr Genet 2015; 5:15-24. [PMID: 27617138 DOI: 10.1055/s-0035-1557109] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/21/2015] [Indexed: 12/26/2022]
Abstract
Nephrotic syndrome (NS) is a common pediatric kidney disease and is defined as massive proteinuria, hypoalbuminemia, and edema. Dysfunction of the glomerular filtration barrier, which is made up of endothelial cells, glomerular basement membrane, and visceral epithelial cells known as podocytes, is evident in children with NS. While most children have steroid-responsive nephrotic syndrome (SSNS), approximately 20% have steroid-resistant nephrotic syndrome (SRNS) and are at risk for progressive kidney dysfunction. While the cause of SSNS is still not well understood, there has been an explosion of research into the genetic causes of SRNS in the past 15 years. More than 30 proteins regulating the function of the glomerular filtration barrier have been associated with SRNS including podocyte slit diaphragm proteins, podocyte actin cytoskeletal proteins, mitochondrial proteins, adhesion and glomerular basement membrane proteins, transcription factors, and others. A genetic cause of SRNS can be found in approximately 70% of infants presenting in the first 3 months of life and 50% of infants presenting between 4 and 12 months, with much lower likelihood for older patients. Identification of the underlying genetic etiology of SRNS is important in children because it allows for counseling of other family members who may be at risk, predicts risk of recurrent disease after kidney transplant, and predicts response to immunosuppressive therapy. Correlations between genetic mutation and clinical phenotype as well as genetic risk factors for SSNS and SRNS are reviewed in this article.
Collapse
Affiliation(s)
- Michelle N Rheault
- Division of Nephrology, University of Minnesota Masonic Children's Hospital, Minneapolis, Minnesota, United States
| | - Rasheed A Gbadegesin
- Division of Nephrology and Center for Human Genetics, Duke University Medical Center, Durham, North Carolina, United States
| |
Collapse
|
45
|
Cil O, Besbas N, Duzova A, Topaloglu R, Peco-Antić A, Korkmaz E, Ozaltin F. Genetic abnormalities and prognosis in patients with congenital and infantile nephrotic syndrome. Pediatr Nephrol 2015; 30:1279-87. [PMID: 25720465 DOI: 10.1007/s00467-015-3058-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 01/08/2015] [Accepted: 01/19/2015] [Indexed: 11/24/2022]
Abstract
BACKGROUND Congenital nephrotic syndrome (CNS) and infantile nephrotic syndrome (INS) are caused primarily by mutations in genes that encode structural and regulatory proteins of the glomerular filtration barrier. The aim of this study was to determine genotype-phenotype correlations and prognosis in patients with CNS and INS. METHODS NPHS1, NPHS2, LAMB2 and the eighth and ninth exons of WT1 were sequenced in 80 and 22 patients with CNS and INS, respectively. Genotype-phenotype correlations and survival were evaluated. RESULTS Causative mutations were identified in 64.7 % of patients, of which NPHS1 mutations were the most common (37.4 %). The mutation detection rate was twofold higher in CNS patients than in INS patients (72.5 vs. 36.2 %). The most commonly mutated gene in CNS patients was NPHS1 (46.3 %) versus NPHS2 (13.6 %) and WT1 (13.6 %) in INS patients. NPHS2 mutations, female patients with NPHS1 mutations, and NPHS1 mutations affecting the transmembrane or intracellular domains of nephrin were associated with longer survival. CONCLUSIONS Based on our present findings, the likelihood of identification of a genetic cause decreases with increasing age at diagnosis. The underlying genetic abnormality should be identified as early as possible, as this knowledge will facilitate clinicians in their prognostic prediction and enable patients to receive appropriate genetic counseling.
Collapse
Affiliation(s)
- Onur Cil
- Department of Pediatric Nephrology, Hacettepe University, Ankara, Turkey
| | | | | | | | | | | | | |
Collapse
|
46
|
Lu L, Sun XM, Yin Y, Huang YF, Wang M, Wan H, Wei LB, Xiao W. The amino acid mutations of the podocin in proteinuria: a meta-analysis. Ren Fail 2015. [PMID: 26211502 DOI: 10.3109/0886022x.2015.1067129] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
While many previous studies have reported an association between the single-nucleotide polymorphisms (SNPs) of the podocin and proteinuria occurred, a conclusive relationship has not been defined in every oligoallelic state of amino acid (AA) mutations in podocin. In this study, we performed a meta-analysis of the published data to investigate the impact of the oligoallelic AA mutations of the podocin on proteinuria; a total 16 AA mutations were investigated for oligoallelic pathogenicity. Despite significant heterogeneity within some of the comparisons, the results revealed significantly higher risks of proteinuria in early-onset (onset age <16) individuals for five mutations (P118L, R138Q, R168H, V180M, and V260E), and in all onset ages individuals for five mutations (R138Q, G140X, R229Q, V260E, and V290M) compared to non-variant individuals. We also tested the steroid response in individuals with R229Q and E237Q. No statistically significant differences in the two mutations carrier rate were observed between steroid resistance patients and controls. No AA mutation was selected for meta-analysis on the recurrence of proteinuria after renal transplantation as lack of control data. In conclusion, our meta-analysis tested the pathogenicity of the oligoallelic AA mutations in podocin and suggested the potential causative mutations, and the alleles showing an association with protein susceptibility. The sensitivity and specificity of each causative mutation are pending further testing.
Collapse
Affiliation(s)
- Lu Lu
- a School of Traditional Chinese Medicine, Southern Medical University , Guangzhou , China
| | - Xiao-ming Sun
- a School of Traditional Chinese Medicine, Southern Medical University , Guangzhou , China
| | - Yi Yin
- a School of Traditional Chinese Medicine, Southern Medical University , Guangzhou , China .,b Department of Nephrology , Southern Medical University TCM-Integrated Hospital , Guangzhou , China .,c Department of Traditional Chinese Medicine , ZhuJiang Hospital, Southern Medical University , Guangzhou , China , and
| | - Yan-feng Huang
- a School of Traditional Chinese Medicine, Southern Medical University , Guangzhou , China .,b Department of Nephrology , Southern Medical University TCM-Integrated Hospital , Guangzhou , China
| | - Ming Wang
- c Department of Traditional Chinese Medicine , ZhuJiang Hospital, Southern Medical University , Guangzhou , China , and
| | - Heng Wan
- d Department of Endocrinology , The Third Affiliated Hospital, Southern Medical University , Guangzhou , China
| | - Lian-Bo Wei
- a School of Traditional Chinese Medicine, Southern Medical University , Guangzhou , China .,b Department of Nephrology , Southern Medical University TCM-Integrated Hospital , Guangzhou , China .,c Department of Traditional Chinese Medicine , ZhuJiang Hospital, Southern Medical University , Guangzhou , China , and
| | - Wei Xiao
- a School of Traditional Chinese Medicine, Southern Medical University , Guangzhou , China
| |
Collapse
|
47
|
Phelan PJ, Hall G, Wigfall D, Foreman J, Nagaraj S, Malone AF, Winn MP, Howell DN, Gbadegesin R. Variability in phenotype induced by the podocin variant R229Q plus a single pathogenic mutation. Clin Kidney J 2015; 8:538-42. [PMID: 26413278 PMCID: PMC4581382 DOI: 10.1093/ckj/sfv063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 06/29/2015] [Indexed: 01/23/2023] Open
Abstract
Background Mutations in podocin (NPHS2) are the most common cause of childhood onset autosomal recessive steroid-resistant nephrotic syndrome (SRNS). The disease is characterized by early-onset proteinuria, resistance to immunosuppressive therapy and rapid progression to end-stage renal disease. Compound heterozygous changes involving the podocin variant R229Q combined with another pathogenic mutation have been associated with a mild phenotype with disease onset often in adulthood. Methods We screened 19 families with early-onset SRNS for mutations in NPHS2 and WT1 and identified four disease-causing mutations (three in NPHS2 and one in WT1) prior to planned whole-exome sequencing. Results We describe two families with three individuals presenting in childhood who are compound heterozygous for R229Q and one other pathogenic NPHS2 mutation, either L327F or A297V. One child presented at age 4 years (A297V plus R229Q) and the other two at age 13 (L327F plus R229Q), one with steadily deteriorating renal function. Conclusions These cases highlight the phenotypic variability associated with the NPHS2 R229Q variant plus pathogenic mutation. Individuals may present with early aggressive disease.
Collapse
Affiliation(s)
- Paul J Phelan
- Duke Molecular Physiology Institute , Duke University , Durham, NC , USA ; Division of Nephrology, Department of Medicine , Duke University Medical Center , Durham, NC , USA ; Department of Nephrology , Royal Infirmary of Edinburgh, NHS Lothian , Edinburgh , UK
| | - Gentzon Hall
- Duke Molecular Physiology Institute , Duke University , Durham, NC , USA ; Division of Nephrology, Department of Medicine , Duke University Medical Center , Durham, NC , USA
| | - Delbert Wigfall
- Department of Nephrology , Royal Infirmary of Edinburgh, NHS Lothian , Edinburgh , UK
| | - John Foreman
- Department of Nephrology , Royal Infirmary of Edinburgh, NHS Lothian , Edinburgh , UK
| | - Shashi Nagaraj
- Department of Nephrology , Royal Infirmary of Edinburgh, NHS Lothian , Edinburgh , UK
| | - Andrew F Malone
- Duke Molecular Physiology Institute , Duke University , Durham, NC , USA ; Division of Nephrology, Department of Medicine , Duke University Medical Center , Durham, NC , USA
| | - Michelle P Winn
- Duke Molecular Physiology Institute , Duke University , Durham, NC , USA ; Division of Nephrology, Department of Medicine , Duke University Medical Center , Durham, NC , USA
| | - David N Howell
- Department of Pathology , Duke University Medical Center , Durham, NC , USA
| | - Rasheed Gbadegesin
- Duke Molecular Physiology Institute , Duke University , Durham, NC , USA ; Department of Pediatrics , Duke University Medical Center , Durham, NC , USA
| |
Collapse
|
48
|
Stefanou C, Pieri M, Savva I, Georgiou G, Pierides A, Voskarides K, Deltas C. Co-Inheritance of Functional Podocin Variants with Heterozygous Collagen IV Mutations Predisposes to Renal Failure. Nephron Clin Pract 2015; 130:200-12. [PMID: 26138234 DOI: 10.1159/000432406] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/16/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS A subset of patients who present with proteinuria and are diagnosed with focal segmental glomerulosclerosis (FSGS) have inherited heterozygous COL4A3/A4 mutations and are also diagnosed with thin basement membrane nephropathy (TBMN-OMIM: 141200). Two studies showed that co-inheritance of NPHS2-p.Arg229Gln, a podocin variant, may increase the risk for proteinuria and renal function decline. METHODS We hypothesized that additional podocin variants may exert a similar effect. We studied genetically a well-characterized Cypriot TBMN patient cohort by re-sequencing the NPHS2 coding region. We also performed functional studies in cell culture experiments, investigating the interaction of podocin variants with itself and with nephrin. RESULTS Potentially disease-modifying podocin variants were searched for by analyzing NPHS2 in 35 'severe' TBMN patients. One non-synonymous variant, p.Glu237Gln, was detected. Both variants, p.Arg229Gln and p.Glu237Gln, were tested in a larger cohort of 122 TBMN patients, who were categorized as 'mild' or 'severe' based on the presence of microscopic hematuria alone or combined with chronic renal failure and/or proteinuria. Seven 'severe' patients carried either of the 2 variants; none was present in the 'mild' patients (p = 0.05, Pearson χ(2)). The 7 carriers belong in 2 families segregating mutation COL4A3-p.Gly1334Glu. Inheritance of the wild-type (WT) and mutant alleles correlated with the phenotype (combined concordance probability 0.003). Immunofluorescence (IF) experiments after dual co-transfection of WT and mutant podocin suggested altered co-localization of mutant homodimers. IF experiments after co-transfection of WT podocin and nephrin showed normal membrane localization, while both podocin variants interfered with normal trafficking, demonstrating perinuclear staining. Immunoprecipitation experiments showed stronger binding of mutant podocin to WT podocin or nephrin. CONCLUSION The results support the hypothesis that certain hypomorphic podocin variants may act as adverse genetic modifiers when co-inherited with COL4A3/A4 mutations, thus predisposing to FSGS and severe kidney function decline.
Collapse
Affiliation(s)
- Charalambos Stefanou
- Molecular Medicine Research Center and Laboratory of Molecular and Medical Genetics, Department of Biological Sciences, University of Cyprus, Nicosia, Cyprus
| | | | | | | | | | | | | |
Collapse
|
49
|
Sadowski CE, Lovric S, Ashraf S, Pabst WL, Gee HY, Kohl S, Engelmann S, Vega-Warner V, Fang H, Halbritter J, Somers MJ, Tan W, Shril S, Fessi I, Lifton RP, Bockenhauer D, El-Desoky S, Kari JA, Zenker M, Kemper MJ, Mueller D, Fathy HM, Soliman NA, Hildebrandt F. A single-gene cause in 29.5% of cases of steroid-resistant nephrotic syndrome. J Am Soc Nephrol 2015; 26:1279-89. [PMID: 25349199 PMCID: PMC4446877 DOI: 10.1681/asn.2014050489] [Citation(s) in RCA: 437] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/10/2014] [Indexed: 01/15/2023] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is the second most frequent cause of ESRD in the first two decades of life. Effective treatment is lacking. First insights into disease mechanisms came from identification of single-gene causes of SRNS. However, the frequency of single-gene causation and its age distribution in large cohorts are unknown. We performed exon sequencing of NPHS2 and WT1 for 1783 unrelated, international families with SRNS. We then examined all patients by microfluidic multiplex PCR and next-generation sequencing for all 27 genes known to cause SRNS if mutated. We detected a single-gene cause in 29.5% (526 of 1783) of families with SRNS that manifested before 25 years of age. The fraction of families in whom a single-gene cause was identified inversely correlated with age of onset. Within clinically relevant age groups, the fraction of families with detection of the single-gene cause was as follows: onset in the first 3 months of life (69.4%), between 4 and 12 months old (49.7%), between 1 and 6 years old (25.3%), between 7 and 12 years old (17.8%), and between 13 and 18 years old (10.8%). For PLCE1, specific mutations correlated with age of onset. Notably, 1% of individuals carried mutations in genes that function within the coenzyme Q10 biosynthesis pathway, suggesting that SRNS may be treatable in these individuals. Our study results should facilitate molecular genetic diagnostics of SRNS, etiologic classification for therapeutic studies, generation of genotype-phenotype correlations, and the identification of individuals in whom a targeted treatment for SRNS may be available.
Collapse
Affiliation(s)
- Carolin E Sadowski
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Svjetlana Lovric
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shazia Ashraf
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Werner L Pabst
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Heon Yung Gee
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stefan Kohl
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Susanne Engelmann
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Virginia Vega-Warner
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, Michigan
| | - Humphrey Fang
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jan Halbritter
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Michael J Somers
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Weizhen Tan
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Shirlee Shril
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Inès Fessi
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard P Lifton
- Department of Genetics and Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut
| | - Detlef Bockenhauer
- Institute of Child Health, University College London, London, United Kingdom
| | - Sherif El-Desoky
- Pediatric Nephrology Unit, King Abdulaziz University Hospital, Jeddah, Kingdom of Saudi Arabia
| | - Jameela A Kari
- Pediatric Nephrology Unit, King Abdulaziz University Hospital, Jeddah, Kingdom of Saudi Arabia
| | - Martin Zenker
- Department of Human Genetics, Otto von Guericke University, Magdeburg, Germany
| | - Markus J Kemper
- Department of Pediatrics, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Mueller
- Department of Pediatric Nephrology, Medical Faculty of the Charité, Berlin, Germany
| | - Hanan M Fathy
- The Pediatric Nephrology Unit, Alexandria University, Alexandria, Egypt
| | - Neveen A Soliman
- Department of Pediatrics, Center of Pediatric Nephrology & Transplantation, Kasr Al Ainy School of Medicine, Cairo University, Cairo, Egypt; Egyptian Group for Orphan Renal Diseases, Cairo, Egypt; and
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts; Howard Hughes Medical Institute, Chevy Chase, Maryland
| |
Collapse
|
50
|
Abstract
Primary disease recurrence after renal transplantation is mainly diagnosed by examination of biopsy samples, but can also be associated with clinical symptoms. In some patients, recurrence can lead to graft loss (7-8% of all graft losses). Primary disease recurrence is generally associated with a high risk of graft loss in patients with focal segmental glomerulosclerosis, membranous proliferative glomerulonephritis, primary hyperoxaluria or atypical haemolytic uraemic syndrome. By contrast, disease recurrence is associated with a limited risk of graft loss in patients with IgA nephropathy, renal involvement associated with Henoch-Schönlein purpura, antineutrophil cytoplasmic antibody-associated glomerulonephritis or lupus nephritis. The presence of systemic diseases that affect the kidneys, such as sickle cell anaemia and diabetes mellitus, also increases the risk of delayed graft loss. This Review provides an overview of the epidemiology, pathophysiology and management of primary disease recurrence in paediatric renal graft recipients, and describes the overall effect on graft survival of each of the primary diseases listed above. With appropriate management, few paediatric patients should be excluded from renal transplantation programmes because of an increased risk of recurrence.
Collapse
|