1
|
Marquez J, O’Sullivan L, Squire AE, Ryan GL, Debiec KE, Amies Oelschlager AM, Adam MP. Case Report: a novel variant in WT1 leads to focal segmental glomerulosclerosis and uterovaginal anomalies through exon skipping. FRONTIERS IN NEPHROLOGY 2025; 5:1542475. [PMID: 40235736 PMCID: PMC11997443 DOI: 10.3389/fneph.2025.1542475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 02/28/2025] [Indexed: 04/17/2025]
Abstract
Background Podocytopathies are a varied set of renal diseases in which podocytes are unable to perform their typical filtration function within the glomerulus. This typically leads to edema, proteinuria, and hypoalbuminemia early in life. Among podocytopathies, focal segmental glomerulosclerosis (FSGS) is characterized by histology demonstrating segmental and focal sclerosis of the glomerular tuft. FSGS affects an estimated 1-20 per one million individuals and leads to significant morbidity and mortality related to renal failure. While FSGS can be attributed to many causes, such as drug reactions and infections, underlying pathogenic genetic variants play an increasingly well-recognized role in this disease. Case A 38-year-old 46,XX female patient of self-reported Cambodian ancestry was evaluated due to her history of atypical uterovaginal morphology. She had a history of hypertension and nephrotic range proteinuria that was diagnosed early in adulthood. A kidney biopsy at that time revealed FSGS. Following worsening renal function and subsequent end-stage renal disease (ESRD), she underwent a kidney transplant at 33 years of age. After kidney transplant, she presented with hematocolpos and was found to have distal vaginal atresia and an arcuate uterus. She underwent vaginoplasty and then had regular menses. She was noted to have persistently elevated follicle stimulating hormone levels, consistent with primary ovarian insufficiency, but with normal anti-Müllerian hormone levels. Assessment of her family history was suggestive of other individuals in her family with similar renal disease and uterine differences. Genetic analysis identified a WT1 variant (c.1338A>C; p. =) of uncertain significance that is also present in her similarly affected mother. To help clarify the potential impact of this variant, we completed a mini-gene assay to detect in vitro splicing changes in the presence of the WT1 variant sequence uncovered in this individual. This demonstrated resultant aberrant splicing that further supports the pathogenicity of the uncovered variant for this individual. Conclusions To our knowledge, this represents the first case of a podocytopathy with co-occurring uterovaginal anomalies due to exon skipping in WT1. The patient exhibited a severe course of chronic kidney dysfunction requiring a kidney transplant. Clinical RNA sequencing to clarify variants impacting splicing remains challenging due to tissue- specific gene expression for genes such as WT1, thus, research-based assays may be beneficial to understand the consequence of rare or previously uncharacterized variants.
Collapse
Affiliation(s)
- Jonathan Marquez
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Lauren O’Sullivan
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Audrey E. Squire
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Ginny L. Ryan
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Washington, Seattle, WA, United States
| | - Katherine E. Debiec
- Department of Obstetrics and Gynecology, Division of Pediatric and Adolescent Gynecology, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| | - Anne-Marie Amies Oelschlager
- Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology and Infertility, University of Washington, Seattle, WA, United States
| | - Margaret P. Adam
- Department of Pediatrics, Division of Genetic Medicine, University of Washington and Seattle Children’s Hospital, Seattle, WA, United States
| |
Collapse
|
2
|
Zhang X, Tian H, Lu C, Xie SP, Ma JS, Chen HZ, Tang DE, Dai Y, Yan Q, Xian W. Long non-coding RNA Growth Arrest Specific 5 Regulates the Podocyte Function in Nephrotic Syndrome Development via microRNA-144-5p/Phosphatase And Tensin Homolog. Nephrology (Carlton) 2025; 30:e70024. [PMID: 40156076 DOI: 10.1111/nep.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
AIM This research examined the role and possible regulatory mechanisms of lncRNA GAS5 in the occurrence and progression of primary nephrotic syndrome (PNS) to provide biomarkers for early screening of PNS in the clinic. METHODS RT-qPCR was employed to assess the expression levels of GAS5 and miR-144-5p. ROC analysis was conducted to evaluate their predictive capabilities for PNS. The interaction between GAS5 and miR-144-5p was confirmed using a dual-luciferase assay. Following this, GAS5 overexpression plasmids, along with co-transfected plasmids, were introduced into podocytes to examine their impact on the inflammatory factors, oxidative stress index, cell proliferation and apoptosis. Furthermore, we performed GO and KEGG enrichment analyses, along with PPI analysis, on the target genes of miR-144-5p to speculate on its potential functions and to identify critical genes. RESULTS The expression levels of GAS5 were decreased while miR-144-5p levels were elevated in PNS patients. The diagnostic approach of serum GAS5 combined with miR-144-5p improved the accuracy of identification. GAS5 was observed to inhibit inflammation and oxidative stress responses and the apoptosis of MPC-5 cells, and enhance cell proliferation. However, the overexpression of miR-144-5p counteracted the effect of GAS5 on podocyte function. Enrichment analysis suggested the miR-144-5p target genes could affect podocyte structure, homeostasis and cell growth. PTEN and STAT3 are identified as critical regulatory targets. CONCLUSION The sponging effect of GAS5 on miR-144-5p caused changes in PTEN mRNA expression and could potentially prevent or mitigate PNS. GAS5 is expected to become a potential target for treating PNS.
Collapse
Affiliation(s)
- Xinyi Zhang
- Department of Infectious Diseases, Frist Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People's Republic of China
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Huan Tian
- Clinical Laboratory, Ezhou Central Hospital, Ezhou, People's Republic of China
| | - Chang Lu
- Department of the Organ Transplantation, 924th Hospital of Joint Logistic Support Force of PLA, Guilin, People's Republic of China
| | - Shen-Ping Xie
- Department of the Organ Transplantation, 924th Hospital of Joint Logistic Support Force of PLA, Guilin, People's Republic of China
| | - Jing-Sheng Ma
- Department of the Organ Transplantation, 924th Hospital of Joint Logistic Support Force of PLA, Guilin, People's Republic of China
| | - Huai-Zhou Chen
- Department of the Organ Transplantation, 924th Hospital of Joint Logistic Support Force of PLA, Guilin, People's Republic of China
| | - Dong-E Tang
- Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, Shenzhen Engineering Research Center of Autoimmune Disease, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Yong Dai
- The First Affiliated Hospital, School of Medicine, Anhui University of Science and Technology, Huainan, People's Republic of China
| | - Qiang Yan
- Department of the Organ Transplantation, 924th Hospital of Joint Logistic Support Force of PLA, Guilin, People's Republic of China
| | - Wei Xian
- Immunology Department, The Second People's Hospital of Henan Province, Zhengzhou, People's Republic of China
| |
Collapse
|
3
|
Nalubega R, Batte A, Kiguli S. Prevalence and predictors of stunting in children and adolescents aged 1-18 years with nephrotic syndrome attending Mulago Hospital, Uganda. BMC Nephrol 2025; 26:110. [PMID: 40033302 DOI: 10.1186/s12882-025-04025-2] [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: 05/31/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Nephrotic syndrome is the predominant glomerulopathy in children worldwide, particularly in low-income countries. One of the key complications of nephrotic syndrome is stunting. Stunting is the most prevalent form of undernutrition globally; which leads to early and long-term consequences. In this study, we determined the prevalence and predictors of stunting among children and adolescents with nephrotic syndrome at a tertiary nephrology clinic in Uganda. METHODS Between February and August 2022, we conducted a cross-sectional study that enrolled children and adolescents aged 1 to 18 years with nephrotic syndrome. Participants had been undergoing steroid treatment for a minimum of three months and were registered at the paediatric renal clinic of Mulago National Referral Hospital in Kampala, Uganda. Medical history, physical examination and anthropometric assessment were conducted on the enrolled children. The World Health Organisation (WHO) growth reference standards were used to evaluate stunting in the enrolled children. Multivariable logistic regression analysis was performed to determine independent predictors of stunting and a p-value < 0.05 was considered statistically significant. RESULTS Ninety-four participants were enrolled, with a median age (IQR) at diagnosis of six years (IQR 3-9). Among the participants, 48 (51.1%) were male. The prevalence of stunting was observed in 15 (15.9%) participants (95% confidence interval [CI]: 15.88 - 16.04). Regarding severity, 12 (12.8%) participants were moderately stunted, and 3 (3.2%) were severely stunted. Participants with persistent proteinuria exhibited higher odds of stunting than those without. (OR: 4.11, 95% CI: 1.05 - 15.98, p < 0.041). CONCLUSIONS There is a high prevalence of stunting among children with nephrotic syndrome, particularly among those with on-going proteinuria. This underscores the importance of regular growth monitoring and screening for early identification and eventual management of stunting among children receiving care for nephrotic syndrome. Providing nutritional counselling and other interventions is thus crucial in addressing stunting among this specific group of children and adolescents.
Collapse
Affiliation(s)
- Raihanah Nalubega
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda.
| | - Anthony Batte
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| | - Sarah Kiguli
- Department of Paediatrics and Child Health, School of Medicine, College of Health Sciences, Makerere University, P.O. Box 7072, Kampala, Uganda
| |
Collapse
|
4
|
Kochuthakidiyel Suresh P, Venkatachalapathy Y, Ekambaram S, Sangeetha, Manoj M, C.D M. Cytochrome P450 3A gene family and medication in childhood nephrotic syndrome: An update. Glob Med Genet 2025; 12:100009. [PMID: 39925446 PMCID: PMC11800309 DOI: 10.1016/j.gmg.2024.100009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 02/11/2025] Open
Abstract
Background Nephrotic syndrome (NS) is a renal disease characterized by excessive proteinuria (greater than 3.5 g/dl per 24 h), which results in hypoalbuminemia and leads to hyperlipidemia, edema, and various complications. NS patients typically respond to standard steroid treatment (prednisolone) and are classified as having steroid-sensitive nephrotic syndrome (SSNS). However, patients who do not respond to steroid therapy after 4 weeks are referred to as having steroid-resistant nephrotic syndrome (SRNS). The unequal response to steroid treatment in nephrotic syndrome involved many factors, including genetic, medication, and kidney diseases. The CYP3A gene family is predominantly involved in the metabolism of medications used in the treatment of NS. Methodology A systematic literature review was conducted from January 2014 to June 2024 using an extensive electronic search of data related to pediatric nephrotic syndrome and the CYP gene family, including associated polymorphisms. Through this review, we systematically analyze factors that affect the metabolism of medications targeting the CYP3A gene family (including steroidal and non-steroidal drugs) commonly used in the treatment of NS and its comorbidities. Conclusion Studies have correlated the relationship between polymorphisms in the CYP3A gene family and medication in NS, with 90 % of the research focusing primarily on post-kidney transplant NS patients. Many studies have reported a correlation between CYP3A gene family polymorphisms and increased tacrolimus (TAC) dosage.
Collapse
Affiliation(s)
| | - Yogalakshmi Venkatachalapathy
- Sri Ramachandra Institute of Higher Education and Research, Department of Human Genetics, Chennai, Tamil Nadu 600116, India
| | - Sudha Ekambaram
- Apollo Children’s Hospital, Department of Pediatric Nephrology, 600006, India
| | - Sangeetha
- Sri Ramachandra Institute of Higher Education and Research, Department of Paediatric Medicine, Chennai, Tamil Nadu 600116, India
| | - Megha Manoj
- Sri Ramachandra Institute of Higher Education and Research, Department of Bioinformatics, Chennai, Tamil Nadu 600116, India
| | - MohanaPriya C.D
- Sri Ramachandra Institute of Higher Education and Research, Department of Human Genetics, Chennai, Tamil Nadu 600116, India
| |
Collapse
|
5
|
Zerkowitz E, Gellermann J, Beckus J, Holle J, Kempf C, Bufler P, Müller D, Thumfart J, Klämbt V. Outcomes and prognostic factors in childhood-onset steroid-resistant nephrotic syndrome: a retrospective single-center study. Pediatr Nephrol 2025:10.1007/s00467-025-06705-5. [PMID: 40021511 DOI: 10.1007/s00467-025-06705-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Steroid-resistant nephrotic syndrome (SRNS) is the second leading cause of chronic kidney disease (CKD) in childhood. It represents a heterogeneous group of diseases with variable kidney outcomes that are still challenging to predict. In this study, our main objective is to describe predictive factors of remission states and kidney survival comparing genetic and non-genetic SRNS. METHODS We conducted a retrospective analysis of 65 pediatric patients with SRNS treated at the pediatric outpatient clinic in Berlin between 2000 and 2023. Clinical characteristics, laboratory findings, and treatment strategies were systematically collected at multiple time points. Outcomes were defined by remission status, kidney survival (CKD stage I-IV), or progression to CKD stage V. Statistical analyses included univariate and multivariate logistic and Cox regression models adjusted for monogenic SRNS to identify predictors of remission and kidney survival. RESULTS The median age of onset was 4.0 years, with a male predominance of 57%. Patients were followed for a median of 5.9 years. At the last follow-up, 26 patients achieved complete remission, 12 achieved partial remission, and 27 showed no remission. Kidney survival rates at 5 and 10 years were 71% and 56%, respectively. High initial nephrotic-range proteinuria, confirmed genetic diagnoses, reduced eGFR, and hypoalbuminemia at 3-month and 1-year follow-ups were identified as negative predictive factors for complete or partial remission. These factors also correlated strongly with an elevated risk of progression to CKD stage V. CONCLUSION Our findings highlight additional prognostic factors influencing remission status and long-term kidney survival in pediatric SRNS, emphasizing the value of detailed early time-point analyses.
Collapse
Affiliation(s)
- Emil Zerkowitz
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Jutta Gellermann
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Juliane Beckus
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Johannes Holle
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Caroline Kempf
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Dominik Müller
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Julia Thumfart
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany
| | - Verena Klämbt
- Department of Pediatric Gastroenterology, Nephrology and Metabolic Diseases, Charité Universitätsmedizin, Berlin, Berlin, Germany.
- Berlin Institute of Health, BIH Charité Clinician Scientist Program, Berlin, Germany.
| |
Collapse
|
6
|
Milovanova A, Ananin P, Vashurina T, Zrobok O, Dmitrienko S, Ryaposova A, Tsygina E, Pushkov A, Zhanin I, Chudakova D, Asanov A, Shchagina O, Polyakov A, Fisenko A, Savostyanov K, Tsygin A. Genetic and Clinical Features of Schimke Immuno-Osseous Dysplasia: Single-Centre Retrospective Study of 21 Unrelated Paediatric Patients over a Period of 20 Years. Int J Mol Sci 2025; 26:1744. [PMID: 40004207 PMCID: PMC11855709 DOI: 10.3390/ijms26041744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Schimke immuno-osseous dysplasia (SIOD) is a hereditary autosomal-recessive multi-system disorder with early mortality. It has variable clinical presentations, mainly characterised by disproportional short stature, steroid-resistant nephrotic syndrome, spondyloepiphyseal dysplasia, and T-cell immunodeficiency. In the majority of cases, SIOD is caused by pathogenic sequence variants (PSVs) in the SMARCAL1 gene that encodes protein involved in chromatin remodelling. SIOD is an ultra-rare condition, with an incidence of ~1 per 1-3 million live births; data on its genetic and clinical features are scarce. We conducted a retrospective study of 21 paediatric patients with SIOD diagnosed in our centre during the years 2003-2023. The most common extra-renal clinical features were short stature, osseous dysplasia, multiple stigmas, and leukopenia. Proteinuria of varying severity was observed in 16 cases. The five-year overall survival rate (OS) was 89% (95% CI 77-100%), and the ten-year OS was 10%. Next-generation sequencing (NGS) revealed the following PSVs in SMARCAL1 in 19 patients: c.355_500del, c.2542G>T, c.2290C>T, c.2562del, c.2533_2534del, c.1582A>C, c.1933C>T, c.1010T>C, c.1736C>T, c.2070dup, c.2551A>T, c.2149_2150dup, c.939delC, and c.1451T>A; the most common was c.2542G>T, resulting in premature translation termination (p.E848*), and it was found in 14 patients either in a homozygous (four patients) or compound-heterozygous (10 patients) state. According to microsatellite analysis, it is a "founder mutation" in Russia.
Collapse
Affiliation(s)
- Anastasiia Milovanova
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Petr Ananin
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Tatiana Vashurina
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Olga Zrobok
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Svetlana Dmitrienko
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Alla Ryaposova
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Elena Tsygina
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Alexander Pushkov
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Ilya Zhanin
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Daria Chudakova
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Aliy Asanov
- Department of Medical Genetics, N.V. Sklifosovsky Institute of Clinical Medicine, I.M. Sechenov First Moscow State Medical University, 115552 Moscow, Russia
| | - Olga Shchagina
- Research Centre for Medical Genetics, 115522 Moscow, Russia
| | | | - Andrey Fisenko
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Kirill Savostyanov
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| | - Alexey Tsygin
- Federal State Autonomous Institution “National Medical Research Center of Children’s Health” of the Ministry of Health of the Russian Federation, 119991 Moscow, Russia
| |
Collapse
|
7
|
Hengel FE, Dehde S, Yilmaz A, Bayazit AK, Ozaltin F, Paripovic D, Emma F, Ronco P, Vivarelli M, Hogan J, Schaefer F, Tomas NM, Huber TB. Anti-nephrin autoantibodies in steroid-resistant nephrotic syndrome may inform treatment strategy. Kidney Int 2025:S0085-2538(25)00082-1. [PMID: 39922375 DOI: 10.1016/j.kint.2025.01.019] [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: 11/21/2024] [Revised: 01/06/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
INTRODUCTION Autoantibodies against the podocyte protein nephrin were recently identified in a pediatric cohort primarily comprising steroid-sensitive (SSNS) and steroid-dependent (SDNS) nephrotic syndrome (NS). However, their prevalence across all NS subtypes, particularly in steroid-resistant nephrotic syndrome (SRNS), and their relation to therapy response need to be determined to advance pathophysiological understanding and refine treatment strategies. METHODS A multicenter cohort study measuring anti-nephrin autoantibodies in samples from children with SSNS, SDNS, nongenetic and genetic SRNS was conducted. RESULTS Sixty-nine of 101 (68%) patients with SSNS, 19 of 67 (28%) patients with SDNS, 14 of 103 patients (14%) with non-genetic SRNS, and 1 of 62 patients (2%) with genetic SRNS were positive for anti-nephrin autoantibodies. The prevalence of anti-nephrin autoantibodies increased with presence of active disease in cases of SSNS and SDNS. Within the group of non-genetic SRNS patients with active disease, anti-nephrin positivity was found in 13 of 74 (18%) patients responding to intensified immunosuppression compared to none of 17 patients with multidrug-resistant SRNS. CONCLUSIONS The prevalence of anti-nephrin antibodies is substantially higher in children with steroid responsive NS than in those with SRNS, suggesting that anti-nephrin antibodies primarily drive SSNS/SDNS. In contrast, NS due to podocyte gene mutations is primarily genotype-caused. Anti-nephrin autoantibodies may serve as a positive prognostic marker in pediatric NS, indicating a favorable response to immunosuppressive therapy.
Collapse
Affiliation(s)
- Felicitas E Hengel
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Silke Dehde
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alev Yilmaz
- Pediatric Nephrology Department, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Türkiye
| | - Aysun K Bayazit
- Department of Pediatric Nephrology, Cukurova University, Adana, Türkiye
| | - Fatih Ozaltin
- Department of Pediatric Nephrology, Hacettepe University, Ankara, Türkiye; Department of Bioinformatics, Hacettepe University, Ankara, Türkiye; Nephrogenetics Laboratory, Department of Pediatric Nephrology, Hacettepe University, Ankara, Türkiye; Center for Genomics and Rare Diseases, Hacettepe University, Ankara, Türkiye
| | - Dusan Paripovic
- Department of Pediatric Nephrology, University Children's Hospital, Belgrade, Serbia
| | - Francesco Emma
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Pierre Ronco
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche S 1155, Paris, France; Division of Nephrology, Centre Hospitalier du Mans, Le Mans, France
| | - Marina Vivarelli
- Laboratory of Nephrology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Julien Hogan
- Pediatric Nephrology Department, Robert Debré Hospital, Assistance Publique Hôpitaux de Paris, Paris, France
| | - Franz Schaefer
- Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.
| | - Nicola M Tomas
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany; Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
8
|
Nagano C, Nozu K. A review of the genetic background in complicated WT1-related disorders. Clin Exp Nephrol 2025; 29:1-9. [PMID: 39002031 PMCID: PMC11807054 DOI: 10.1007/s10157-024-02539-x] [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: 05/07/2024] [Accepted: 07/05/2024] [Indexed: 07/15/2024]
Abstract
The Wilms tumor 1 (WT1) gene was first identified in 1990 as a strong candidate for conferring a predisposition to Wilms tumor. The WT1 protein has four zinc finger structures (DNA binding domain) at the C-terminus, which bind to transcriptional regulatory sequences on DNA, and acts as a transcription factor. WT1 is expressed during kidney development and regulates differentiation, and is also expressed in glomerular epithelial cells after birth to maintain the structure of podocytes. WT1-related disorders are a group of conditions associated with an aberrant or absent copy of the WT1 gene. This group of conditions encompasses a wide phenotypic spectrum that includes Denys-Drash syndrome (DDS), Frasier syndrome (FS), Wilms-aniridia-genitourinary-mental retardation syndrome, and isolated manifestations of nephropathy or Wilms tumor. The genotype-phenotype correlation is becoming clearer: patients with missense variants in DNA binding sites including C2H2 sites manifest DDS and develop early-onset and rapidly developing end-stage kidney disease. A deeper understanding of the genotype-phenotype correlation has also been obtained in DDS, but no such correlation has been observed in FS. The incidence of Wilms tumor is higher in patients with DDS and exon-truncating variants than in those with non-truncating variants. Here, we briefly describe the genetic background of this highly complicated WT1-related disorders.
Collapse
Affiliation(s)
- China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan.
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-Cho, Chuo-Ku, Kobe, 650-0017, Japan
| |
Collapse
|
9
|
Lim C, Lim RS, Choo J, Leow EH, Chan GC, Zhang Y, Ng JL, Chin HL, Tan ES, Goh J, Gandhi N, Ng YH, Than M, Ganesan I, Chong SL, Yap C, Chao SM, Cham B, Kam S, Lim JY, Mok I, Tan HZ, Kwek JL, Lee TL, Wang Z, Goh SM, Lim R, Yeo SC, Teo BW, Da Y, Matchar D, Ng KH. Clinical Implementation of Nephrologist-Led Genomic Testing for Glomerular Diseases in Singapore: Rationale and Protocol. Am J Nephrol 2024; 56:158-171. [PMID: 39626636 PMCID: PMC11975324 DOI: 10.1159/000542942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 12/02/2024] [Indexed: 01/14/2025]
Abstract
INTRODUCTION The early diagnosis and appropriate treatment of monogenic glomerular diseases can reduce kidney failure, avoid unnecessary investigations such as kidney biopsies and ineffective treatment with immunosuppressants, guide transplant decisions, and inform the genetic risks of their family members. Yet, genetic testing for kidney disease is underutilized in Singapore. We aimed to implement a nephrologist-led genetic service and evaluate the acceptance, adoption, utility, and cost-effectiveness of genetic testing for monogenic glomerular disease in Singapore. METHODS We will perform a prospective, multi-centre, type II hybrid effectiveness-implementation study with a post-design to evaluate both implementation and clinical outcomes of nephrologist-led genetic testing for suspected genetic glomerular kidney diseases. The multi-disciplinary implementation team will train "genetic nephrologists" to provide pre- and post-test counselling, order targeted exome panel sequencing for suspected glomerular kidney diseases (persistent microscopic haematuria and/or albuminuria or proteinuria in the absence of known causes, steroid-resistant primary nephrotic syndrome, apparent familial IgA nephropathy, or chronic kidney disease with no apparent cause), and interpret genetic test results; create workflows for patient referral, evaluation and management, and discuss genetic results at regular genomic board meetings. The outcomes are acceptance, appropriateness and adoption among patients and nephrologists, utility (proportion of patients who received genetic testing and have a confirmed diagnosis of genetic glomerular disease), and cost-effectiveness. CONCLUSION This study will create and evaluate a nephrologist-led genetic service, develop an efficient variant curation process, and inform future recommendations on the optimal referral and genetic testing strategy for monogenic glomerular disease in Singapore. This will facilitate the future mainstreaming of genetic testing that will enable precision medicine in kidney care.
Collapse
Affiliation(s)
- Cynthia Lim
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ru Sin Lim
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Jason Choo
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Esther Huimin Leow
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Gek Cher Chan
- Department of Medicine, Nephrology, National University Hospital, Singapore, Singapore
| | - Yaochun Zhang
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Jun Li Ng
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Hui-Lin Chin
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Ee Shien Tan
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Jeannette Goh
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Naline Gandhi
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
| | - Yong Hong Ng
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Mya Than
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| | - Indra Ganesan
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Siew Le Chong
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Celeste Yap
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Sing Ming Chao
- Department of Paediatrics, Nephrology, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Breana Cham
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Sylvia Kam
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Jiin Ying Lim
- Department of Paediatrics, Genetics, KK Women’s and Children’s Hospital, Singapore, Singapore
| | - Irene Mok
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Hui Zhuan Tan
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Jia Liang Kwek
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Tung Lin Lee
- Department of Renal Medicine, Medicine, Singapore General Hospital, SingHealth Duke-NUS Academic Medical Center, Singapore, Singapore
| | - Ziyin Wang
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Su Mein Goh
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Regina Lim
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - See Cheng Yeo
- Department of Renal Medicine, Tan Tock Seng Hospital, Singapore, Singapore
| | - Boon Wee Teo
- Department of Medicine, Nephrology, National University of Singapore, Singapore, Singapore
| | - Yi Da
- Department of Medicine, Nephrology, National University of Singapore, Singapore, Singapore
| | - David Matchar
- Program in Health Services and Systems Research, Duke-NUS Medical School, Singapore, Singapore
- Department of Medicine (General Internal Medicine), Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Kar Hui Ng
- Department of Paediatrics, National University of Singapore, Singapore, Singapore
| |
Collapse
|
10
|
Li S, Hu M, He C, Sun Y, Huang W, Lei F, Liu Y, Huang Z, Meng Y, Liu W, Lei X, Dong Y, Lin Z, Huang C, Zhao R, Qin Y. A multicenter study investigating the genetic analysis of childhood steroid-resistant nephrotic syndrome: Variants in COL4A5 may not be coincidental. PLoS One 2024; 19:e0304864. [PMID: 39625990 PMCID: PMC11614205 DOI: 10.1371/journal.pone.0304864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 05/20/2024] [Indexed: 12/06/2024] Open
Abstract
This study aimed to discuss the pathogenic hereditary factors of children with steroid-resistant nephrotic syndrome (SRNS) in Guangxi, China. We recruited 89 patients with SRNS or infantile NS from five major pediatric nephrology centers in Guangxi, and conducted a retrospective analysis of clinical data. Whole-exome sequencing analysis was also performed on all patients. The risk of progression to chronic kidney disease (CKD) was assessed using the Kaplan-Meier method and Cox proportional hazards model. The study included 69 male and 20 female participants from 86 distinct families, with the median age of disease onset being 48 months (interquartile range: 24-93). Overall, 24.7% had a family history of SRNS, whereas 13.5% exhibited extra-kidney manifestations. We identified disease-causing variants in 24.7% (22/89) of patients across eight screened genes. The most frequently detected variant was found in COL4A5, followed by NPHS2 (5.6%), NPHS1 (2.2%), PAX2 (2.2%), WT1 (1.1%), LMX1B (1.1%), NUP105 (1.1%), and COL4A6 (1.1%). Twelve of the 26 pathogenic variants were determined to be de novo. Based on gene detection results, pathogenic variants were categorized into two groups: identified and unidentified variants. The identified variant group demonstrated a significant association with positive family history, steroid resistant-style, and response to immune therapy (P<0.001). Patients with the identified genetic variant were approximately ten times more likely to develop CKD (P<0.001) than those in the unidentified group at the last follow-up. Kidney biopsy was performed on 66 patients, and minimal change disease was the most prevalent histopathological diagnosis (29 cases; 32.6%). These findings suggest that children diagnosed with SRNS exhibit a diverse range of genetic alterations. We identified the COL4A5 variant as the predominant genetic abnormality and a low frequency of NPHS1 gene involvement in these children. Gene variants may serve as an independent predictor for SRNS progression to CKD.
Collapse
Affiliation(s)
- Sheng Li
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
- Department of Pediatrics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Miaoyue Hu
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
| | - Chao He
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
- Department of Pediatrics, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Yu Sun
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
| | - Weifang Huang
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
| | - Fengying Lei
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
| | - Yunguang Liu
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi Province, China
| | - Zengpo Huang
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical College for Nationalities, Baise, Guangxi Province, China
| | - Yongqiu Meng
- Department of Pediatrics, Guigang People’s Hospital, Guigang, China
| | - Wenjing Liu
- Department of Pediatrics, Guigang People’s Hospital, Guigang, China
| | - Xianqiang Lei
- Department of Pediatrics, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Yanfang Dong
- Department of Pediatrics, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, China
| | - Zihui Lin
- Department of Pediatrics, Maternity and Child Healthcare of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Chunlin Huang
- Department of Pediatrics, Maternity and Child Healthcare of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Rihong Zhao
- Department of Pediatrics, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Yuanhan Qin
- Department of Pediatrics, The First Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
11
|
Liu PJ, Sayeeda K, Zhuang C, Krendel M. Roles of myosin 1e and the actin cytoskeleton in kidney functions and familial kidney disease. Cytoskeleton (Hoboken) 2024; 81:737-752. [PMID: 38708443 PMCID: PMC11538376 DOI: 10.1002/cm.21861] [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: 12/28/2023] [Revised: 03/21/2024] [Accepted: 04/04/2024] [Indexed: 05/07/2024]
Abstract
Mammalian kidneys are responsible for removing metabolic waste and maintaining fluid and electrolyte homeostasis via selective filtration. One of the proteins closely linked to selective renal filtration is myosin 1e (Myo1e), an actin-dependent molecular motor found in the specialized kidney epithelial cells involved in the assembly and maintenance of the renal filter. Point mutations in the gene encoding Myo1e, MYO1E, have been linked to familial kidney disease, and Myo1e knockout in mice leads to the disruption of selective filtration. In this review, we discuss the role of the actin cytoskeleton in renal filtration, the known and hypothesized functions of Myo1e, and the possible explanations for the impact of MYO1E mutations on renal function.
Collapse
Affiliation(s)
- Pei-Ju Liu
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Kazi Sayeeda
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Cindy Zhuang
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Mira Krendel
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
12
|
Li Q, Yang Z, Zang R, Liu S, Yu L, Wang J, Wang C, Wang X, Sun S. Clinical features and genetic analysis of 15 Chinese children with dent disease. Ren Fail 2024; 46:2349133. [PMID: 38726999 PMCID: PMC11089919 DOI: 10.1080/0886022x.2024.2349133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 04/19/2024] [Indexed: 05/15/2024] Open
Abstract
OBJECTIVE The clinical characteristics, genetic mutation spectrum, treatment strategies and prognoses of 15 children with Dent disease were retrospectively analyzed to improve pediatricians' awareness of and attention to this disease. METHODS We analyzed the clinical and laboratory data of 15 Chinese children with Dent disease who were diagnosed and treated at our hospital between January 2017 and May 2023 and evaluated the expression of the CLCN5 and OCRL1 genes. RESULTS All 15 patients were male and complained of proteinuria, and the incidence of low-molecular-weight proteinuria (LMWP) was 100.0% in both Dent disease 1 (DD1) and Dent disease 2 (DD2) patients. The incidence of hypercalciuria was 58.3% (7/12) and 66.7% (2/3) in DD1 and DD2 patients, respectively. Nephrocalcinosis and nephrolithiasis were found in 16.7% (2/12) and 8.3% (1/12) of DD1 patients, respectively. Renal biopsy revealed focal segmental glomerulosclerosis (FSGS) in 1 patient, minimal change lesion in 5 patients, and small focal acute tubular injury in 1 patient. A total of 11 mutations in the CLCN5 gene were detected, including 3 missense mutations (25.0%, c.1756C > T, c.1166T > G, and c.1618G > A), 5 frameshift mutations (41.7%, c.407delT, c.1702_c.1703insC, c.137delC, c.665_666delGGinsC, and c.2200delG), and 3 nonsense mutations (25.0%, c.776G > A, c.1609C > T, and c.1152G > A). There was no significant difference in age or clinical phenotype among patients with different mutation types (p > 0.05). All three mutations in the OCRL1 gene were missense mutations (c.1477C > T, c.952C > T, and c.198A > G). CONCLUSION Pediatric Dent disease is often misdiagnosed. Protein electrophoresis and genetic testing can help to provide an early and correct diagnosis.
Collapse
Affiliation(s)
- Qian Li
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Zhenle Yang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Ruixian Zang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Suwen Liu
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Lichun Yu
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Jing Wang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Cong Wang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Xiaoyuan Wang
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| | - Shuzhen Sun
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
- Department of Pediatric Nephrology and Rheumatism and Immunology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, P.R. China
| |
Collapse
|
13
|
Lee JX, Tan YJ, Ismail NAS. NPHS Mutations in Pediatric Patients with Congenital and Steroid-Resistant Nephrotic Syndrome. Int J Mol Sci 2024; 25:12275. [PMID: 39596340 PMCID: PMC11594456 DOI: 10.3390/ijms252212275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 11/02/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
NPHS1 and NPHS2 are kidney gene components that encode for nephrin and podocin, respectively. They play a role in the progression of congenital (CNS) and steroid-resistant (SRNS) nephrotic syndrome. Hence, this study aimed to determine the prevalence and renal outcomes of NPHS mutations among pediatric patients with CNS and SRNS. We also aimed to identify potential predictors of NPHS mutations in this patient cohort. Overall, this study included 33 studies involving 2123 patients screened for NPHS1, whereas 2889 patients from 40 studies were screened for NPHS2 mutations. The patients' mean age was 4.9 ± 1 years (ranging from birth to 18 years), and 56% of patients were male (n = 1281). Using the random-effects model, the pooled proportion of NPHS1 mutations among pediatric patients with CNS and SRNS was 0.15 (95% CI 0.09; 0.24, p < 0.001, I2 = 92.0%). The pooled proportion of NPHS2 mutations was slightly lower, at 0.11 (95% CI 0.08; 0.14, p < 0.001, I2 = 73.8%). Among the 18 studies that reported ESRF, the pooled proportion was 0.47 (95% CI 0.34; 0.61, p < 0.001, I2 = 75.4%). Our study showed that the NPHS1 (β = 1.16, p = 0.35) and NPHS2 (β = 5.49, p = 0.08) mutations did not predict ESRF in CNS and SRNS pediatric patients. Nevertheless, patients from the European continent who had the NPHS2 mutation had a significantly higher risk of developing ESRF (p < 0.05, β = 1.3, OR = 7.97, 95% CI 0.30; 2.30) compared to those who had the NPHS1 mutation. We recommend NPHS mutation screening for earlier diagnosis and to avoid unnecessary steroid treatments. More data are needed to better understand the impact of NPHS mutations among pediatric patients with CNS and SRNS.
Collapse
Affiliation(s)
- Jun Xin Lee
- Department of Pediatrics, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Yan Jin Tan
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| | - Noor Akmal Shareela Ismail
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
14
|
Meliambro K, He JC, Campbell KN. Podocyte-targeted therapies - progress and future directions. Nat Rev Nephrol 2024; 20:643-658. [PMID: 38724717 DOI: 10.1038/s41581-024-00843-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2024] [Indexed: 09/14/2024]
Abstract
Podocytes are the key target cells for injury across the spectrum of primary and secondary proteinuric kidney disorders, which account for up to 90% of cases of kidney failure worldwide. Seminal experimental and clinical studies have established a causative link between podocyte depletion and the magnitude of proteinuria in progressive glomerular disease. However, no substantial advances have been made in glomerular disease therapies, and the standard of care for podocytopathies relies on repurposed immunosuppressive drugs. The past two decades have seen a remarkable expansion in understanding of the mechanistic basis of podocyte injury, with prospects increasing for precision-based treatment approaches. Dozens of disease-causing genes with roles in the pathogenesis of clinical podocytopathies have been identified, as well as a number of putative glomerular permeability factors. These achievements, together with the identification of novel targets of podocyte injury, the development of potential approaches to harness the endogenous podocyte regenerative potential of progenitor cell populations, ongoing clinical trials of podocyte-specific pharmacological agents and the development of podocyte-directed drug delivery systems, contribute to an optimistic outlook for the future of glomerular disease therapy.
Collapse
Affiliation(s)
- Kristin Meliambro
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John C He
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kirk N Campbell
- Department of Medicine, Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
15
|
Finn LS. Nephrotic Syndrome Throughout Childhood: Diagnosing Podocytopathies From the Womb to the Dorm. Pediatr Dev Pathol 2024; 27:426-458. [PMID: 38745407 DOI: 10.1177/10935266241242669] [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: 05/16/2024]
Abstract
The etiologies of podocyte dysfunction that lead to pediatric nephrotic syndrome (NS) are vast and vary with age at presentation. The discovery of numerous novel genetic podocytopathies and the evolution of diagnostic technologies has transformed the investigation of steroid-resistant NS while simultaneously promoting the replacement of traditional morphology-based disease classifications with a mechanistic approach. Podocytopathies associated with primary and secondary steroid-resistant NS manifest as diffuse mesangial sclerosis, minimal change disease, focal segmental glomerulosclerosis, and collapsing glomerulopathy. Molecular testing, once an ancillary option, has become a vital component of the clinical investigation and when paired with kidney biopsy findings, provides data that can optimize treatment and prognosis. This review focuses on the causes including selected monogenic defects, clinical phenotypes, histopathologic findings, and age-appropriate differential diagnoses of nephrotic syndrome in the pediatric population with an emphasis on podocytopathies.
Collapse
Affiliation(s)
- Laura S Finn
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at The University of Pennsylvania, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| |
Collapse
|
16
|
Lopez-Gonzalez M, Ariceta G. WT1-related disorders: more than Denys-Drash syndrome. Pediatr Nephrol 2024; 39:2601-2609. [PMID: 38326647 DOI: 10.1007/s00467-024-06302-y] [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: 10/03/2023] [Revised: 01/07/2024] [Accepted: 01/07/2024] [Indexed: 02/09/2024]
Abstract
Historically, specific mutations in WT1 gene have been associated with distinct syndromes based on phenotypic characteristics, including Denys-Drash syndrome (DDS), Frasier syndrome (FS), Meacham syndrome, and WAGR syndrome. DDS is classically defined by the triad of steroid-resistant nephrotic syndrome (SRNS) onset in the first year of life, disorders of sex development (DSD), and a predisposition to Wilms tumor (WT). Currently, a paradigm shift acknowledges a diverse spectrum of presentations beyond traditional syndromic definitions. Consequently, the concept of WT1-related disorders becomes more precise. A genotype-phenotype correlation has been established, emphasizing that the location and type of WT1 mutations significantly influence the clinical presentation, the condition severity, and the chronology of patient manifestations. Individuals presenting with persistent proteinuria, with or without nephrotic syndrome, and varying degrees of kidney dysfunction accompanied by genital malformations should prompt suspicion of WT1 mutations. Recent genetic advances enable a more accurate estimation of malignancy risk in these patients, facilitating a conservative nephron-sparing surgery (NSS) approach in select cases, with a focus on preserving residual kidney function and delaying nephrectomies. Other key management strategies include kidney transplantation and addressing DSD and gonadoblastoma. In summary, recent genetic insights underscore the imperative to implement individualized, integrated, and multidisciplinary management strategies for WT1-related disorders. This approach is pivotal in optimizing patient outcomes and addressing the complexities associated with these diverse clinical manifestations.
Collapse
Affiliation(s)
| | - Gema Ariceta
- Department of Pediatric Nephrology, University Hospital Vall d'Hebron, Barcelona, Spain
- University Autonomous of Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
Platt CJ, Bierzynska A, Ding W, Saleem SA, Koziell A, Saleem MA. Rare heterozygous variants in paediatric steroid resistant nephrotic syndrome - a population-based analysis of their significance. Sci Rep 2024; 14:18568. [PMID: 39127776 DOI: 10.1038/s41598-024-68837-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
Genetic testing in nephrotic syndrome may identify heterozygous predicted-pathogenic variants (HPPVs) in autosomal recessive (AR) genes that are known to cause disease in the homozygous or compound heterozygous state. In such cases, it can be difficult to define the variant's true significance and questions remain about whether a second pathogenic variant has been missed during analysis or whether the variant is an incidental finding. There are now known to be over 70 genes associated with nephrotic syndrome, the majority inherited as an AR trait. Knowledge of whether such HPPVs occur with equal frequency in patients compared to the general population would assist interpretation of their significance. Exome sequencing was performed on 187 Steroid-Resistant Nephrotic Syndrome (SRNS) paediatric patients recruited to a UK rare disease registry plus originating from clinics at Evelina, London. 59 AR podocytopathy linked genes were analysed in each patient and a list of HPPVs created. We compared the frequency of detected HPPVs with a 'control' population from the gnomAD database containing exome data from approximately 50,000 individuals. A bespoke filtering process was used for both patients and controls to predict 'likely pathogenicity' of variants. In total 130 Caucasian SRNS patients were screened across 59 AR genes and 201 rare heterozygous variants were identified. 17/201 (8.5%) were assigned as 'likely pathogenic' (HPPV) using our bespoke filtering method. Comparing each gene in turn, for SRNS patients with a confirmed genetic diagnosis, in 57 of the 59 genes we found no statistically significant difference in the frequency of these HPPVs between patients and controls (In genes ARHGDIA and TP53RK, we identified a significantly higher number of HPPVs in the control population compared with the patients when filtering was performed with 'high stringency' settings only). In the SRNS patients without a genetics diagnosis confirmed, there was no statistically significant difference identified in any gene between patient and control. In children with SRNS, we propose that identification of HPPV in AR podocytopathy linked genes is not necessarily representative of pathogenicity, given that the frequency is similar to that seen in controls for the majority. Whilst this may not exclude the presence of genetic kidney disease, this type of heterozygous variant is unlikely to be causal and each result must be interpreted in its clinical context.
Collapse
Affiliation(s)
- C J Platt
- Bristol Royal Hospital for Children, Bristol, BS2 8NJ, UK.
| | - A Bierzynska
- Bristol Renal, University of Bristol, Bristol, UK
| | - W Ding
- Bristol Renal, University of Bristol, Bristol, UK
| | | | - A Koziell
- King's College and Evelina, London, UK
| | - M A Saleem
- Bristol Renal, University of Bristol, Bristol, UK
| |
Collapse
|
18
|
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
|
19
|
王 英, 何 庆. [Research progress on monogenic inherited glomerular diseases with central nervous system symptoms]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:652-658. [PMID: 38926384 PMCID: PMC11562061 DOI: 10.7499/j.issn.1008-8830.2312054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/15/2024] [Indexed: 06/28/2024]
Abstract
To date, approximately 500 monogenic inherited kidney diseases have been reported, with more than 50 genes associated with the pathogenesis of monogenic isolated or syndromic nephrotic syndrome. Most of these genes are expressed in podocytes of the glomerulus. Neurological symptoms are common extrarenal manifestations of syndromic nephrotic syndrome, and various studies have found connections between podocytes and neurons in terms of morphology and function. This review summarizes the genetic and clinical characteristics of monogenic inherited diseases with concomitant glomerular and central nervous system lesions, aiming to enhance clinicians' understanding of such diseases, recognize the importance of genetic diagnostic techniques for comorbidity screening, and reduce the rates of missed diagnosis and misdiagnosis.
Collapse
|
20
|
Buerger F, Merz LM, Saida K, Yu S, Salmanullah D, Lemberg K, Mertens ND, Mansour B, Kolvenbach CM, Yousef K, Hölzel S, Braun A, Franken GAC, Goncalves KA, Steinsapir A, Endlich N, Schneider R, Shril S, Hildebrandt F. Quantitative phenotyping of Nphs1 knockout mice as a prerequisite for gene replacement studies. Am J Physiol Renal Physiol 2024; 326:F780-F791. [PMID: 38482553 PMCID: PMC11386980 DOI: 10.1152/ajprenal.00412.2023] [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: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/22/2024] Open
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is the second most frequent cause of chronic kidney disease before the age of 25 yr. Nephrin, encoded by NPHS1, localizes to the slit diaphragm of glomerular podocytes and is the predominant structural component of the glomerular filtration barrier. Biallelic variants in NPHS1 can cause congenital nephrotic syndrome of the Finnish type, for which, to date, no causative therapy is available. Recently, adeno-associated virus (AAV) vectors targeting the glomerular podocyte have been assessed as a means for gene replacement therapy. Here, we established quantitative and reproducible phenotyping of a published, conditional Nphs1 knockout mouse model (Nphs1tm1.1Pgarg/J and Nphs2-Cre+) in preparation for a gene replacement study using AAV vectors. Nphs1 knockout mice (Nphs1fl/fl Nphs2-Cre+) exhibited 1) a median survival rate of 18 days (range: from 9 to 43 days; males: 16.5 days and females: 20 days); 2) an average foot process (FP) density of 1.0 FP/µm compared with 2.0 FP/µm in controls and a mean filtration slit density of 2.64 µm/µm2 compared with 4.36 µm/µm2 in controls; 3) a high number of proximal tubular microcysts; 4) the development of proteinuria within the first week of life as evidenced by urine albumin-to-creatinine ratios; and 5) significantly reduced levels of serum albumin and elevated blood urea nitrogen and creatinine levels. For none of these phenotypes, significant differences between sexes in Nphs1 knockout mice were observed. We quantitatively characterized five different phenotypic features of congenital nephrotic syndrome in Nphs1fl/fl Nphs2-Cre+ mice. Our results will facilitate future gene replacement therapy projects by allowing for sensitive detection of even subtle molecular effects.NEW & NOTEWORTHY To evaluate potential, even subtle molecular, therapeutic effects of gene replacement therapy (GRT) in a mouse model, prior rigorous quantifiable and reproducible disease phenotyping is necessary. Here, we, therefore, describe such a phenotyping effort in nephrin (Nphs1) knockout mice to establish the basis for GRT for congenital nephrotic syndrome. We believe that our findings set an important basis for upcoming/ongoing gene therapy approaches in the field of nephrology, especially for monogenic nephrotic syndrome.
Collapse
Affiliation(s)
- Florian Buerger
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- University Children's Hospital, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lea M Merz
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Department of Pediatrics, University Leipzig, Leipzig, Germany
| | - Ken Saida
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Seyoung Yu
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Daanya Salmanullah
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Katharina Lemberg
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Nils D Mertens
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Bshara Mansour
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Caroline M Kolvenbach
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
- Institute of Anatomy, Medical Faculty, University of Bonn, Bonn, Germany
| | - Kirollos Yousef
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Selina Hölzel
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Alina Braun
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Gijs A C Franken
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Kevin A Goncalves
- Deerfield Discovery and Development, Deerfield Management Company, L.P. (Series C), New York, New York, United States
| | - Andrew Steinsapir
- Deerfield Discovery and Development, Deerfield Management Company, L.P. (Series C), New York, New York, United States
| | | | - Ronen Schneider
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Shirlee Shril
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, United States
| |
Collapse
|
21
|
Han Y, Sha H, Yang Y, Yu Z, Zhou L, Wang Y, Yang F, Qiu L, Zhang Y, Zhou J. Mutations in the NUP93, NUP107 and NUP160 genes cause steroid-resistant nephrotic syndrome in Chinese children. Ital J Pediatr 2024; 50:81. [PMID: 38650033 PMCID: PMC11036785 DOI: 10.1186/s13052-024-01656-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 04/07/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND The variants of nucleoporins are extremely rare in hereditary steroid-resistant nephrotic syndrome (SRNS). Most of the patients carrying such variants progress to end stage kidney disease (ESKD) in their childhood. More clinical and genetic data from these patients are needed to characterize their genotype-phenotype relationships and elucidate the role of nucleoporins in SRNS. METHODS Four patients of SRNS carrying biallelic variants in the NUP93, NUP107 and NUP160 genes were presented. The clinical and molecular genetic characteristics of these patients were summarized, and relevant literature was reviewed. RESULTS All four patients in this study were female and initially presented with SRNS. The median age at the onset of the disease was 5.08 years, ranging from 1 to 10.5 years. Among the four patients, three progressed to ESKD at a median age of 7 years, ranging from 1.5 to 10.5 years, while one patient reached stage 3 chronic kidney disease (CKD3). Kidney biopsies revealed focal segmental glomerulosclerosis in three patients. Biallelic variants were detected in NUP93 in one patient, NUP107 in two patients, as well as NUP160 in one patient respectively. Among these variants, five yielded single amino acid substitutions, one led to nonsense mutation causing premature termination of NUP107 translation, one caused a single nucleotide deletion resulting in frameshift and truncation of NUP107. Furthermore, one splicing donor mutation was observed in NUP160. None of these variants had been reported previously. CONCLUSION This report indicates that biallelic variants in NUP93, NUP107 and NUP160 can cause severe early-onset SRNS, which rapidly progresses to ESKD. Moreover, these findings expand the spectrum of phenotypes and genotypes and highlight the importance of next-generation sequencing in elucidating the molecular basis of SRNS and allowing rational treatment for affected individuals.
Collapse
Affiliation(s)
- Yanxinli Han
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Hongyu Sha
- Department of Pharmacy, Yantai Yuhuangding Hospital, Yantai, Shandong Province, 264000, China
| | - Yuan Yang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Zhuowei Yu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Lanqi Zhou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Yi Wang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Fengjie Yang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Liru Qiu
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Yu Zhang
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China
| | - Jianhua Zhou
- Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei province, 430030, China.
| |
Collapse
|
22
|
Badeńska M, Pac M, Badeński A, Rutkowska K, Czubilińska-Łada J, Płoski R, Bohynikova N, Szczepańska M. A Rare De Novo Mutation in the TRIM8 Gene in a 17-Year-Old Boy with Steroid-Resistant Nephrotic Syndrome: Case Report. Int J Mol Sci 2024; 25:4486. [PMID: 38674071 PMCID: PMC11050435 DOI: 10.3390/ijms25084486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Idiopathic nephrotic syndrome is the most common chronic glomerular disease in children. Treatment with steroids is usually successful; however, in a small percentage of patients, steroid resistance is observed. The most frequent histologic kidney feature of steroid-resistant nephrotic syndrome (SRNS) is focal segmental glomerulosclerosis (FSGS). Genetic testing has become a valuable diagnostic tool in defining the etiology of SRNS, leading to the identification of a genetic cause. The TRIM8 gene is expressed in various tissues, including kidney cells and the central nervous system (CNS). An association between a mutation in the TRIM8 gene and an early onset of FSGS has been proposed but is not well described. We present a 17-year-old boy with epilepsy, early mild developmental delay, a low IgG serum level, and proteinuria, secondary to FSGS. A Next-Generation Sequencing (NGS)-based analysis revealed a heterozygous de novo pathogenic variant in the TRIM8 gene (c.1200C>G, p.Tyr400Ter). TRIM8 gene sequencing should be considered in individuals with early onset of FSGS, particularly accompanied by symptoms of cortical dysfunction, such as epilepsy and intellectual disability.
Collapse
Affiliation(s)
- Marta Badeńska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (M.B.); (A.B.)
| | - Małgorzata Pac
- Department of Immunology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.P.); (N.B.)
| | - Andrzej Badeński
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (M.B.); (A.B.)
| | - Karolina Rutkowska
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (K.R.); (R.P.)
| | - Justyna Czubilińska-Łada
- Department of Neonatal Intensive Care and Neonatal Pathology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland;
| | - Rafał Płoski
- Department of Medical Genetics, Medical University of Warsaw, 02-106 Warsaw, Poland; (K.R.); (R.P.)
| | - Nadezda Bohynikova
- Department of Immunology, The Children’s Memorial Health Institute, 04-730 Warsaw, Poland; (M.P.); (N.B.)
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, ul. 3 Maja 13/15, 41-800 Zabrze, Poland; (M.B.); (A.B.)
| |
Collapse
|
23
|
Jankowski M, Daca-Roszak P, Bałasz-Chmielewska I, Ustaszewski A, Żurowska A, Lipska-Ziętkiewicz BS, Ziętkiewicz E. Estimation of the Age of the Kashubian-Specific Pathogenic NPHS2 Variant Responsible for Hereditary Steroid-Resistant Nephrotic Syndrome Points to Its Recent Local Origin. Hum Mutat 2024; 2024:8205102. [PMID: 40225918 PMCID: PMC11918915 DOI: 10.1155/2024/8205102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 02/05/2024] [Accepted: 02/16/2024] [Indexed: 04/15/2025]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is a highly heterogenic kidney disorder resulting from genetic abnormalities or immune system dysfunction affecting the establishment and maintenance of the glomerular filtration barrier. The most common cause of genetic SRNS is biallelic pathogenic variants in NPHS2 gene, especially in individuals with an infantile or childhood onset. The type of the NPHS2 defect implies the course of the disease and the stage of its onset and differs across populations. In a cohort of Polish patients with SRNS, a unique profile of the disease-related NPHS2 variants was identified in patients from northern Poland inhabited by Kashubs, a minority West-Slavic ethnic group known for a local increase of the frequency of several pathogenic variants. Among Kashubian families, the compound heterozygotes c.686G>A/c.1032delT and a single c.1032delT homozygote were the only underlying cause of SRNS. The restricted, Kashubian-only pattern of c.1032delT occurrence, suggesting the founder effect, prompted us to conduct a detailed analysis of its haplotype background to estimate the age of the c.1032delT origin. Eight Kashubian SRNS families were genotyped using the Infinium Global Screening Array-24. The haplotype background analysis was performed using an in-house pipeline designed to solve the phase of the heterozygous genotype data. The age of the c.1032delT mutation was calculated using the gamma method based on the genetic length of ancestral haplotypes shared between two or more individuals carrying this variant. The results of our study indicated a very recent origin of the c.1032delT mutation (~240 years). Genetic screening performed in the general Polish population control corroborates the assumption that the mutation occurred on the specific Kashubian haplotype background. The identification of ancestry-specific Kashubian pathogenic variant can help to develop effective screening and diagnostic strategies as a part of personalized medicine approach in the region.
Collapse
Affiliation(s)
- M. Jankowski
- Department of Biology and Medical Genetics, Medical University of Gdansk, Gdansk, Poland
| | - P. Daca-Roszak
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - I. Bałasz-Chmielewska
- Department of Pediatrics, Nephrology and Hypertension, Medical University of Gdansk, Gdansk, Poland
| | - A. Ustaszewski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| | - A. Żurowska
- Department of Pediatrics, Nephrology and Hypertension, Medical University of Gdansk, Gdansk, Poland
- Centre for Rare Diseases, Medical University of Gdansk, Gdansk, Poland
| | - B. S. Lipska-Ziętkiewicz
- Centre for Rare Diseases, Medical University of Gdansk, Gdansk, Poland
- Clinical Genetics Unit, Department of Biology and Medical Genetics, Medical University of Gdansk, Gdansk, Poland
| | - E. Ziętkiewicz
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
24
|
Zhang L, Zhao F, Ding G, Chen Y, Zhao S, Chen Q, Sha Y, Che R, Huang S, Zheng B, Zhang A. Monogenic Causes Identified in 23.68% of Children with Steroid-Resistant Nephrotic Syndrome: A Single-Centre Study. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:61-68. [PMID: 38322629 PMCID: PMC10843177 DOI: 10.1159/000534853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/22/2023] [Indexed: 02/08/2024]
Abstract
Introduction Steroid-resistant nephrotic syndrome (SRNS) is the second most common cause of end-stage kidney disease in children, mostly associated with focal segmental glomerulosclerosis (FSGS). Advances in genomic science have enabled the identification of causative variants in 20-30% of SRNS patients. Methods We used whole exome sequencing to explore the genetic causes of SRNS in children. Totally, 101 patients with SRNS and 13 patients with nephrotic proteinuria and FSGS were retrospectively enrolled in our hospital between 2018 and 2022. For the known monogenic causes analysis, we generated a known SRNS gene list of 71 genes through reviewing the OMIM database and literature. Results Causative variants were identified in 23.68% of our cohort, and the most frequently mutated genes in our cohort were WT1 (7/27), NPHS1 (3/27), ADCK4 (3/27), and ANLN (2/27). Five patients carried variants in phenocopy genes, including MYH9, MAFB, TTC21B, AGRN, and FAT4. The variant detection rate was the highest in the two subtype groups with congenital nephrotic syndrome and syndromic SRNS. In total, 68.75% of variants we identified were novel and have not been previously reported in the literature. Conclusion Comprehensive genetic analysis is key to realizing the clinical benefits of a genetic diagnosis. We suggest that all children with SRNS undergo genetic testing, especially those with early-onset and extrarenal phenotypes.
Collapse
Affiliation(s)
- Luyan Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Fei Zhao
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Guixia Ding
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Chen
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Sanlong Zhao
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Qiuxia Chen
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Yugen Sha
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Ruochen Che
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Bixia Zheng
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
25
|
Ambarsari CG, Utami DAP, Tandri CC, Satari HI. Comparison of three spot proteinuria measurements for pediatric nephrotic syndrome: based on the International pediatric Nephrology Association 2022 Guidelines. Ren Fail 2023; 45:2253324. [PMID: 37724557 PMCID: PMC10512887 DOI: 10.1080/0886022x.2023.2253324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 08/24/2023] [Indexed: 09/21/2023] Open
Abstract
BACKGROUND Pediatric nephrotic syndrome (NS) requires routine proteinuria monitoring, which is costly and affects patients' quality of life. The gold-standard 24-h urine protein (UP) measurement is challenging in children, and first-morning urine collection requires specific conditions, making it difficult in outpatient settings. Studies have reported comparability of second or random morning urine sample to the first-morning specimen. This study aimed to compare outcomes of random morning proteinuria measurements to 24-h UP and the roles of the urinary protein creatinine ratio (UPCR) and dipstick tests in pediatric NS, based on International Pediatric Nephrology Association (IPNA) 2022 Guidelines. METHOD Twenty-four-hour and morning urine samples were collected from 92 pediatric NS patients. These were subjected to automated analyses for 24-h UP, UPCR, and semi-automated dipstick analysis. A blinded doctor performed manual dipstick analysis. RESULTS UPCR had a stronger correlation with 24-h UP than with automated and manual urine dipstick tests. UPCR had the highest sensitivity and specificity for predicting no remission/relapse and high sensitivity but low specificity for complete remission. The optimal UPCR cutoff for remission was 0.44 mg/mg and for no remission/relapse was 2.08 mg/mg. Automated and manual dipstick tests demonstrated limited sensitivity but high specificity and similar AUC values for remission/relapse. CONCLUSION UPCR was sensitive and specific for diagnosing no remission/relapse and sensitive but not specific for detecting remission. Manual and automated urine dipstick tests were comparable for remission and no remission/relapse detection. This study supports the IPNA 2022 Guidelines, as 2 mg/mg was the optimal UPCR cutoff for no remission/relapse, while for remission the optimal cutoff was 0.4 mg/mg.
Collapse
Affiliation(s)
- Cahyani Gita Ambarsari
- Department of Child Health, Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- School of Medicine, University of Nottingham, Nottingham, United Kingdom
- Medical Technology Cluster, Indonesian Medical Education and Research Institute (IMERI), Universitas Indonesia, Jakarta, Indonesia
| | - Dwi Ambar Prihatining Utami
- Department of Child Health, Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
- Bina Husada Hospital (Member of Mitra Keluarga), Bogor, Indonesia
| | | | - Hindra Irawan Satari
- Department of Child Health, Universitas Indonesia - Cipto Mangunkusumo Hospital, Jakarta, Indonesia
| |
Collapse
|
26
|
Vivarelli M, Gibson K, Sinha A, Boyer O. Childhood nephrotic syndrome. Lancet 2023; 402:809-824. [PMID: 37659779 DOI: 10.1016/s0140-6736(23)01051-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/04/2023] [Accepted: 05/19/2023] [Indexed: 09/04/2023]
Abstract
Idiopathic nephrotic syndrome is the most common glomerular disease in children. Corticosteroids are the cornerstone of its treatment, and steroid response is the main prognostic factor. Most children respond to a cycle of oral steroids, and are defined as having steroid-sensitive nephrotic syndrome. Among the children who do not respond, defined as having steroid-resistant nephrotic syndrome, most respond to second-line immunosuppression, mainly with calcineurin inhibitors, and children in whom a response is not observed are described as multidrug resistant. The pathophysiology of nephrotic syndrome remains elusive. In cases of immune-mediated origin, dysregulation of immune cells and production of circulating factors that damage the glomerular filtration barrier have been described. Conversely, up to a third of cases of steroid-resistant nephrotic syndrome have a monogenic origin. Multidrug resistant nephrotic syndrome often leads to kidney failure and can cause relapse after kidney transplant. Although steroid-sensitive nephrotic syndrome does not affect renal function, most children with steroid-sensitive nephrotic syndrome have a relapsing course that requires repeated steroid cycles with significant side-effects. To minimise morbidity, some patients require steroid-sparing immunosuppressive agents, including levamisole, mycophenolate mofetil, calcineurin inhibitors, anti-CD20 monoclonal antibodies, and cyclophosphamide. Close monitoring and preventive measures are warranted at onset and during relapse to prevent acute complications (eg, hypovolaemia, acute kidney injury, infections, and thrombosis), whereas long-term management requires minimising treatment-related side-effects. A subset of patients have active disease into adulthood.
Collapse
Affiliation(s)
- Marina Vivarelli
- Division of Nephrology, Laboratory of Nephrology, Bambino Gesù Children's Hospital IRCCS, Rome, Italy.
| | - Keisha Gibson
- Division of Nephrology and Hypertension, University of North Carolina Kidney Center, University of North Carolina at Chapel Hill, NC, USA
| | - Aditi Sinha
- Division of Nephrology, Indian Council of Medical Research Center for Advanced Research in Nephrology, Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Olivia Boyer
- Néphrologie Pédiatrique, Centre de Référence Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Hôpital Necker - Enfants Malades, Assistance Publique Hôpitaux de Paris, Inserm U1163, Institut Imagine, Université Paris Cité, Paris, France
| |
Collapse
|
27
|
Saha A, Kapadia SF, Vala KB, Patel HV. Clinical utility of genetic testing in Indian children with kidney diseases. BMC Nephrol 2023; 24:212. [PMID: 37464296 DOI: 10.1186/s12882-023-03240-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 06/11/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Kidney diseases with genetic etiology in children present with an overlapping spectrum of manifestations. We aimed to analyze the clinical utility of genetic testing in the diagnosis and management of suspected genetic kidney diseases in children. METHODS In this retrospective study, children ≤ 18 years in whom a genetic test was ordered were included. Clinical indications for genetic testing were categorized as Glomerular diseases, nephrolithiasis and/or nephrocalcinoses, tubulopathies, cystic kidney diseases, congenital abnormality of kidney and urinary tract, chronic kidney disease of unknown aetiology and others. Clinical exome sequencing was the test of choice. Other genetic tests ordered were sanger sequencing, gene panel, multiplex ligation-dependent probe amplification and karyotyping. The pathogenicity of the genetic variant was interpreted as per the American College of Medical Genetics classification. RESULTS A total of 86 samples were sent for genetic testing from 76 index children, 8 parents and 2 fetuses. A total of 74 variants were reported in 47 genes. Out of 74 variants, 42 were missense, 9 nonsense, 12 frameshifts, 1 indel, 5 affected the splicing regions and 5 were copy number variants. Thirty-two were homozygous, 36 heterozygous and 6 were hemizygous variants. Twenty-four children (31.6%) had pathogenic and 11 (14.5%) had likely pathogenic variants. Twenty-four children (31.6%) had variants of uncertain significance. No variants were reported in 17 children (22.3%). A genetic diagnosis was made in 35 children with an overall yield of 46%. The diagnostic yield was 29.4% for glomerular diseases, 53.8% for tubular disorders, 81% for nephrolithiasis and/or nephrocalcinoses, 60% for cystic kidney diseases and 50% for chronic kidney disease of unknown etiology. Genetic testing made a new diagnosis or changed the diagnosis in 15 children (19.7%). CONCLUSION Nearly half (46%) of the children tested for a genetic disease had a genetic diagnosis. Genetic testing confirmed the clinical diagnoses, changed the clinical diagnoses or made a new diagnosis which helped in personalized management.
Collapse
Affiliation(s)
- Anshuman Saha
- Department of Pediatric Nephrology, Institute of Kidney Diseases and Research Centre, Gujarat University of Transplantation Sciences, Ahmedabad, India.
| | - Shahenaz F Kapadia
- Department of Pediatric Nephrology, Institute of Kidney Diseases and Research Centre, Gujarat University of Transplantation Sciences, Ahmedabad, India
| | - Kinnari B Vala
- Department of Pediatric Nephrology, Institute of Kidney Diseases and Research Centre, Gujarat University of Transplantation Sciences, Ahmedabad, India
| | - Himanshu V Patel
- Department of Pediatric Nephrology, Institute of Kidney Diseases and Research Centre, Gujarat University of Transplantation Sciences, Ahmedabad, India
- Department of Nephrology, Institute of Kidney Diseases and Research Centre, Gujarat University of Transplantation Sciences, Ahmedabad, India
| |
Collapse
|
28
|
Williams AE, Esezobor CI, Lane BM, Gbadegesin RA. Hiding in plain sight: genetics of childhood steroid-resistant nephrotic syndrome in Sub-Saharan Africa. Pediatr Nephrol 2023; 38:2003-2012. [PMID: 36459247 PMCID: PMC10416081 DOI: 10.1007/s00467-022-05831-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 12/03/2022]
Abstract
Steroid-resistant nephrotic syndrome (SRNS) is the most severe form of childhood nephrotic syndrome with an increased risk of progression to chronic kidney disease stage 5. Research endeavors to date have identified more than 80 genes that are associated with SRNS. Most of these genes regulate the structure and function of the podocyte, the visceral epithelial cells of the glomerulus. Although individuals of African ancestry have the highest prevalence of SRNS, especially those from Sub-Saharan Africa (SSA), with rates as high as 30-40% of all cases of nephrotic syndrome, studies focusing on the characterization and understanding of the genetic basis of SRNS in the region are negligible compared with Europe and North America. Therefore, it remains unclear if some of the variants in SRNS genes that are deemed pathogenic for SRNS are truly disease causing, and if the leading causes of monogenic nephrotic syndrome in other populations are the same for children in SSA with SRNS. Other implications of this lack of genetic data for SRNS in the region include the exclusion of children from the region from clinical trials aimed at identifying potential novel therapeutic agents for this severe form of nephrotic syndrome. This review underlines a need for concerted efforts to advance the genetic basis of SRNS in children in SSA. Such endeavors will complement global efforts at understanding the genetic basis of nephrotic syndrome.
Collapse
Affiliation(s)
- Anna Elizabeth Williams
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, 27710, USA
| | - Christopher I Esezobor
- Department of Pediatrics, Faculty of Clinical Sciences, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Brandon M Lane
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, 27710, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA
| | - Rasheed A Gbadegesin
- Department of Pediatrics, Division of Nephrology, Duke University Medical Center, Durham, NC, 27710, USA.
- Duke Molecular Physiology Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
29
|
Elshafey SA, Thabet MAEH, Elwafa RAHA, Schneider R, Shril S, Buerger F, Hildebrandt F, Fathy HM. Genetic stratification reveals COL4A variants and spontaneous remission in Egyptian children with proteinuria in the first 2 years of life. Acta Paediatr 2023; 112:1324-1332. [PMID: 36847718 PMCID: PMC10175230 DOI: 10.1111/apa.16732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/23/2023] [Accepted: 02/24/2023] [Indexed: 03/01/2023]
Abstract
AIM The earlier the onset of proteinuria, the higher the incidence of genetic forms. Therefore, we aimed to analyse the spectrum of monogenic proteinuria in Egyptian children presenting at age <2 years. METHODS The results of 27-gene panel or whole-exome sequencing were correlated with phenotype and treatment outcomes in 54 patients from 45 families. RESULTS Disease-causing variants were identified in 29/45 (64.4%) families. Mutations often occurred in three podocytopathy genes: NPHS1, NPHS2 and PLCE1 (19 families). Some showed extrarenal manifestations. Additionally, mutations were detected in 10 other genes, including novel variants of OSGEP, SGPL1 and SYNPO2. COL4A variants phenocopied isolated steroid-resistant nephrotic syndrome (2/29 families, 6.9%). NPHS2 M1L was the single most common genetic finding beyond the age of 3 months (4/18 families, 22.2%). Biopsy results did not correlate with genotypes (n = 30). On renin-angiotensin-aldosterone system antagonists alone, partial and complete remission occurred in 3/24 (12.5%) patients with monogenic proteinuria each, whereas 6.3% (1/16) achieved complete remission on immunosuppression. CONCLUSION Genotyping is mandatory to avoid biopsies and immunosuppression when proteinuria presents at age <2 years. Even with such a presentation, COL4A genes should be included. NPHS2 M1L was prevalent in Egyptian children (4 months-2 years) with proteinuria, demonstrating precision diagnostic utility.
Collapse
Affiliation(s)
- Samar Atef Elshafey
- Pediatric Nephrology Unit, Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | | | | | - Ronen Schneider
- Division of Nephrology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shirlee Shril
- Division of Nephrology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Florian Buerger
- Division of Nephrology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Friedhelm Hildebrandt
- Division of Nephrology, Department of Pediatrics, Boston Children’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Hanan M Fathy
- Pediatric Nephrology Unit, Department of Pediatrics, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
30
|
Malakasioti G, Iancu D, Milovanova A, Tsygin A, Horinouchi T, Nagano C, Nozu K, Kamei K, Fujinaga S, Iijima K, Sinha R, Basu B, Morello W, Montini G, Waters A, Boyer O, Yıldırım ZY, Yel S, Dursun İ, McCarthy HJ, Vivarelli M, Prikhodina L, Besouw MTP, Chan EYH, Huang W, Kemper MJ, Loos S, Prestidge C, Wong W, Zlatanova G, Ehren R, Weber LT, Chehade H, Hooman N, Tkaczyk M, Stańczyk M, Miligkos M, Tullus K. A multicenter retrospective study of calcineurin inhibitors in nephrotic syndrome secondary to podocyte gene variants. Kidney Int 2023; 103:962-972. [PMID: 36898413 DOI: 10.1016/j.kint.2023.02.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 02/04/2023] [Accepted: 02/16/2023] [Indexed: 03/12/2023]
Abstract
While 44-83% of children with steroid-resistant nephrotic syndrome (SRNS) without a proven genetic cause respond to treatment with a calcineurin inhibitor (CNI), current guidelines recommend against the use of immunosuppression in monogenic SRNS. This is despite existing evidence suggesting that remission with CNI treatment is possible and can improve prognosis in some cases of monogenic SRNS. Herein, our retrospective study assessed response frequency, predictors of response and kidney function outcomes among children with monogenic SRNS treated with a CNI for at least three months. Data from 203 cases (age 0-18 years) were collected from 37 pediatric nephrology centers. Variant pathogenicity was reviewed by a geneticist, and 122 patients with a pathogenic and 19 with a possible pathogenic genotype were included in the analysis. After six months of treatment and at last visit, 27.6% and 22.5% of all patients respectively, demonstrated partial or full response. Achievement of at least partial response at six months of treatment conferred a significant reduction in kidney failure risk at last follow-up compared to no response (hazard ratio [95% confidence interval] 0.25, [0.10-0.62]). Moreover, risk of kidney failure was significantly lower when only those with a follow-up longer than two years were considered (hazard ratio 0.35, [0.14-0.91]). Higher serum albumin level at CNI initiation was the only factor related to increased likelihood of significant remission at six months (odds ratio [95% confidence interval] 1.16, [1.08-1.24]). Thus, our findings justify a treatment trial with a CNI also in children with monogenic SRNS.
Collapse
Affiliation(s)
| | - Daniela Iancu
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | | | - Alexey Tsygin
- National Medical Research Centre of Children's Health, Moscow, Russia
| | - Tomoko Horinouchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Koichi Kamei
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Shuichiro Fujinaga
- Division of Nephrology, Saitama Children's Medical Center, Saitama, Japan
| | | | - Rajiv Sinha
- Division of Pediatric Nephrology, Institute of Child Health, Kolkata, India
| | - Biswanath Basu
- Division of Pediatric Nephrology, Department of Pediatrics, Nil Ratan Sircar Medical College and Hospital, Kolkata, India
| | - William Morello
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca'Granda, Ospedale Maggiore Policlinico di Milano, Milan, Italy
| | - Giovanni Montini
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca'Granda, Ospedale Maggiore Policlinico di Milano, Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Aoife Waters
- Department of Paediatric Nephrology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| | - Olivia Boyer
- Department of Pediatric Nephrology, Hôpital Necker Enfants Malades, AP-HP and Université de Paris, Paris, France
| | - Zeynep Yürük Yıldırım
- Division of Pediatric Nephrology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sibel Yel
- Department of Pediatric Nephrology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - İsmail Dursun
- Department of Pediatric Nephrology, Erciyes University Medical Faculty, Kayseri, Turkey
| | - Hugh J McCarthy
- Department of Nephrology, Sydney Children's Hospitals Network & Department of Child and Adolescent Health, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Marina Vivarelli
- Division of Nephrology and Dialysis, Department of Pediatric Subspecialties, Bambino Gesù Pediatric Hospital IRCCS, Rome, Italy
| | - Larisa Prikhodina
- Veltishev Research & Clinical Institute for Pediatrics, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Martine T P Besouw
- Department of Pediatric Nephrology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | | | - Wenyan Huang
- Department of Nephrology and Rheumatology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Markus J Kemper
- University Children's Hospital, University Medical Center-Hamburg-Eppendorf, Hamburg, Germany
| | - Sebastian Loos
- University Children's Hospital, University Medical Center-Hamburg-Eppendorf, Hamburg, Germany
| | | | - William Wong
- Starship Children's Hospital, Auckland, New Zealand
| | - Galia Zlatanova
- Department of Nephrology and Dialysis University Pediatric Hospital "Prof. Ivan Mitev", Department of Pediatrics Medical University, Sofia, Bulgaria
| | - Rasmus Ehren
- Pediatric Nephrology, Children's and Adolescents' Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Lutz T Weber
- Pediatric Nephrology, Children's and Adolescents' Hospital, University Hospital of Cologne, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Hassib Chehade
- Division of Pediatrics and DOHaD Lab, CHUV-UNIL, Lausanne, Switzerland
| | - Nakysa Hooman
- Aliasghar Clinical Research Development Center, Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marcin Tkaczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Medical University of Lodz, Lodz, Poland
| | - Małgorzata Stańczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Medical University of Lodz, Lodz, Poland
| | - Michael Miligkos
- Second Department of Pediatrics, University of Athens School of Medicine and P. & A. Kyriakou Children's Hospital, Athens, Greece
| | - Kjell Tullus
- Department of Paediatric Nephrology, Great Ormond Street Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
31
|
Arslan Z, Webb H, Ashton E, Foxler B, Tullus K, Waters A, Bockenhauer D. Mendelian steroid resistant nephrotic syndrome in childhood: is it as common as reported? Pediatr Nephrol 2023; 38:1051-1056. [PMID: 35802272 DOI: 10.1007/s00467-022-05569-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 03/18/2022] [Accepted: 03/31/2022] [Indexed: 11/28/2022]
Abstract
BACKGROUND Primary steroid resistant nephrotic syndrome (SRNS) is thought to have either genetic or immune-mediated aetiology. Knowing which children to screen for genetic causes can be difficult. Several studies have described the prevalence of genetic causes of primary SRNS to be between 30 and 40%, but these may reflect a selection bias for genetic testing in children with congenital, infantile, syndromic or familial NS and thus may overestimate the true prevalence in a routine clinical setting. METHODS Retrospective electronic patient record analysis was undertaken of all children with non-syndromic SRNS and presentation beyond the first year of life, followed at our centre between 2005 and 2020. RESULTS Of the 49 children who met the inclusion criteria, 5 (10%) had causative variants identified, predominantly in NPHS2. None responded to immunosuppression. Of the 44 (90%) who had no genetic cause identified, 33 (75%) had complete or partial remission after commencing second-line immunosuppression and 67% of these had eGFR > 90 ml/min/1.73 m2 at last clinical follow-up. Of the children who did not respond to immunosuppression, 64% progressed to kidney failure. CONCLUSIONS In our cohort of children with non-syndromic primary SRNS and presentation beyond the first year of life, we report a prevalence of detectable causative genetic variants of 10%. Those with identified genetic cause were significantly (p = 0.003) less likely to respond to immunosuppression and more likely (p = 0.026) to progress to chronic kidney disease. Understanding the genetics along with response to immunosuppression informs management in this cohort of patients and variant interpretation. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Zainab Arslan
- Department of Paediatric Nephrology, Great Ormond Street Hospital, London, UK.
| | - Hazel Webb
- Department of Paediatric Nephrology, Great Ormond Street Hospital, London, UK
| | - Emma Ashton
- NE Thames Regional Genetics Laboratory, GOSH NHS Foundation Trust, London, UK
| | - Becky Foxler
- Department of Paediatric Nephrology, Great Ormond Street Hospital, London, UK
| | - Kjell Tullus
- Department of Paediatric Nephrology, Great Ormond Street Hospital, London, UK
| | - Aoife Waters
- Department of Paediatric Nephrology, Great Ormond Street Hospital, London, UK
| | - Detlef Bockenhauer
- Department of Paediatric Nephrology, Great Ormond Street Hospital, London, UK.,Department of Renal Medicine, University College London, London, UK
| |
Collapse
|
32
|
Mishra OP, Sidar M, Batra VV, Prasad R, Singh A, Abhinay A, Mishra A, Yadav AK. Outcomes of children with idiopathic steroid resistant nephrotic syndrome: a single centre observational study. J Bras Nefrol 2023; 45:199-209. [PMID: 36179014 PMCID: PMC10627125 DOI: 10.1590/2175-8239-jbn-2022-0073en] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 06/20/2022] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Idiopathic steroid resistant nephrotic syndrome (SRNS) has variable outcomes in children. The primary objective of the present study was to assess the cumulative remission rate and the secondary objectives were to assess factors affecting the remission status, kidney function survival, and adverse effects of medications. METHODS One hundred fourteen patients with SRNS were included. Calcineurin inhibitor-based treatment protocol along with prednisolone and angiotensin-converting enzyme inhibitor were used, and patients were followed over 5 years. RESULTS Median age was 4.5 years; 53.5% of cases were between 1 to 5 years of age. Sixty-two patients (54.4%) were at initial stage and 52 (45.6%) were at a late SRNS stage. Median eGFRcr was 83.5 mL/min/1.73m2 at presentation. Of the 110 patients, 63 (57.3%) achieved remission [complete remission 30 (27.3%), partial remission 33 (30%)], and 47 (42.7%) had no remission. Kidney function survival was 87.3% and 14 cases (12.7%) had progression to CKD (G3-8, G4-3, G5-1, and G5D-2). Median duration of follow up was 36 months (IQR 24, 60). Age of onset, cyclosporine/tacrolimus, eGFRcr, and histopathology (MCD/FSGS) did not affect remission. Similarly, remission status in addition to age of onset, drug protocol, and histopathology did not significantly affect kidney function during a period of 5 years. Hypertension, cushingoid facies, short stature, cataract, and obesity were observed in 37.7, 29.8, 25.5, 17.5, and 0.7% of cases, respectively. CONCLUSION About half of the cases achieved remission. Age of onset of disease, cyclosporine/tacrolimus use, and histopathological lesion neither affected remission status nor short-term kidney function survival in SRNS.
Collapse
Affiliation(s)
- Om P. Mishra
- Banaras Hindu University, Institute of Medical Sciences, Department
of Pediatrics, Division of Pediatric Nephrology, Varanasi, India
| | - Minketan Sidar
- Banaras Hindu University, Institute of Medical Sciences, Department
of Pediatrics, Division of Pediatric Nephrology, Varanasi, India
| | - Vineeta V. Batra
- G. B. Pant Institute of Post Graduate Medical Education &
Research, Department of Pathology, New Delhi, India
| | - Rajniti Prasad
- Banaras Hindu University, Institute of Medical Sciences, Department
of Pediatrics, Division of Pediatric Nephrology, Varanasi, India
| | - Ankur Singh
- Banaras Hindu University, Institute of Medical Sciences, Department
of Pediatrics, Division of Pediatric Nephrology, Varanasi, India
| | - Abhishek Abhinay
- Banaras Hindu University, Institute of Medical Sciences, Department
of Pediatrics, Division of Pediatric Nephrology, Varanasi, India
| | - Akash Mishra
- Jawahar Lal Institute of Postgraduate Medical Education and
Research, Department of Biostatistics, Puducherry, India
| | - Ashish K. Yadav
- Banaras Hindu University, Institute of Medical Sciences, Center of
Biostatistics, Varanasi, India
| |
Collapse
|
33
|
Becherucci F, Landini S, Palazzo V, Cirillo L, Raglianti V, Lugli G, Tiberi L, Dirupo E, Bellelli S, Mazzierli T, Lomi J, Ravaglia F, Sansavini G, Allinovi M, Giannese D, Somma C, Spatoliatore G, Vergani D, Artuso R, Rosati A, Cirami C, Dattolo PC, Campolo G, De Chiara L, Papi L, Vaglio A, Lazzeri E, Anders HJ, Mazzinghi B, Romagnani P. A Clinical Workflow for Cost-Saving High-Rate Diagnosis of Genetic Kidney Diseases. J Am Soc Nephrol 2023; 34:706-720. [PMID: 36753701 PMCID: PMC10103218 DOI: 10.1681/asn.0000000000000076] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 12/19/2022] [Indexed: 01/22/2023] Open
Abstract
SIGNIFICANCE STATEMENT To optimize the diagnosis of genetic kidney disorders in a cost-effective manner, we developed a workflow based on referral criteria for in-person evaluation at a tertiary center, whole-exome sequencing, reverse phenotyping, and multidisciplinary board analysis. This workflow reached a diagnostic rate of 67%, with 48% confirming and 19% modifying the suspected clinical diagnosis. We obtained a genetic diagnosis in 64% of children and 70% of adults. A modeled cost analysis demonstrated that early genetic testing saves 20% of costs per patient. Real cost analysis on a representative sample of 66 patients demonstrated an actual cost reduction of 41%. This workflow demonstrates feasibility, performance, and economic effect for the diagnosis of genetic kidney diseases in a real-world setting. BACKGROUND Whole-exome sequencing (WES) increases the diagnostic rate of genetic kidney disorders, but accessibility, interpretation of results, and costs limit use in daily practice. METHODS Univariable analysis of a historical cohort of 392 patients who underwent WES for kidney diseases showed that resistance to treatments, familial history of kidney disease, extrarenal involvement, congenital abnormalities of the kidney and urinary tract and CKD stage ≥G2, two or more cysts per kidney on ultrasound, persistent hyperechoic kidneys or nephrocalcinosis on ultrasound, and persistent metabolic abnormalities were most predictive for genetic diagnosis. We prospectively applied these criteria to select patients in a network of nephrology centers, followed by centralized genetic diagnosis by WES, reverse phenotyping, and multidisciplinary board discussion. RESULTS We applied this multistep workflow to 476 patients with eight clinical categories (podocytopathies, collagenopathies, CKD of unknown origin, tubulopathies, ciliopathies, congenital anomalies of the kidney and urinary tract, syndromic CKD, metabolic kidney disorders), obtaining genetic diagnosis for 319 of 476 patients (67.0%) (95% in 21 patients with disease onset during the fetal period or at birth, 64% in 298 pediatric patients, and 70% in 156 adult patients). The suspected clinical diagnosis was confirmed in 48% of the 476 patients and modified in 19%. A modeled cost analysis showed that application of this workflow saved 20% of costs per patient when performed at the beginning of the diagnostic process. Real cost analysis of 66 patients randomly selected from all categories showed actual cost reduction of 41%. CONCLUSIONS A diagnostic workflow for genetic kidney diseases that includes WES is cost-saving, especially if implemented early, and is feasible in a real-world setting.
Collapse
Affiliation(s)
- Francesca Becherucci
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Samuela Landini
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Viviana Palazzo
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Luigi Cirillo
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Valentina Raglianti
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Gianmarco Lugli
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Lucia Tiberi
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Elia Dirupo
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | | | - Tommaso Mazzierli
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Jacopo Lomi
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | | | - Giulia Sansavini
- Nephrology and Dialysis Unit, Santo Stefano Hospital, Prato, Italy
| | - Marco Allinovi
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence, Italy
| | | | - Chiara Somma
- Nephrology Unit Florence 1, Santa Maria Annunziata Hospital, Bagno a Ripoli, Florence, Italy
| | - Giuseppe Spatoliatore
- Nephrology and Dialysis Unit, San Giovanni di Dio Hospital, AUSL Toscana Centro, Florence, Italy
| | - Debora Vergani
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Rosangela Artuso
- Medical Genetics Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Alberto Rosati
- Nephrology and Dialysis Unit, San Giovanni di Dio Hospital, AUSL Toscana Centro, Florence, Italy
| | - Calogero Cirami
- Nephrology, Dialysis and Transplantation Unit, Careggi University Hospital, Florence, Italy
| | - Pietro Claudio Dattolo
- Nephrology Unit Florence 1, Santa Maria Annunziata Hospital, Bagno a Ripoli, Florence, Italy
| | - Gesualdo Campolo
- Nephrology and Dialysis Unit, Santo Stefano Hospital, Prato, Italy
| | - Letizia De Chiara
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Laura Papi
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Augusto Vaglio
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Elena Lazzeri
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| | - Hans-Joachim Anders
- Division of Nephrology, Medizinische Klinik and Poliklinik IV, Klinikum der LMU München, Munich, Germany
| | - Benedetta Mazzinghi
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
| | - Paola Romagnani
- Nephrology and Dialysis Unit, Meyer Children's Hospital IRCCS, Florence, Italy
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio,” University of Florence, Florence, Italy
| |
Collapse
|
34
|
Tastemel Ozturk T, Canpolat N, Saygili S, Bayrakci US, Soylemezoglu O, Ozaltin F, Topaloglu R. A rare cause of nephrotic syndrome-sphingosine-1-phosphate lyase (SGPL1) deficiency: 6 cases and a review of the literature. Pediatr Nephrol 2023; 38:711-719. [PMID: 35748945 DOI: 10.1007/s00467-022-05656-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND Recently, recessive mutations in SGPL1 (sphingosine-1-phosphate lyase), which encodes the final enzyme of sphingolipid metabolism, have been reported to cause steroid-resistant nephrotic syndrome, adrenal insufficiency, and many other organ/system involvements. We aimed to determine the clinical and genetic characteristics, and outcomes in patients with SGPL1 mutations. METHODS The study included 6 patients with bi-allelic SGPL1 mutation. Clinical, genetic, and laboratory characteristics, and outcomes of the patients were evaluated retrospectively. We also reviewed previously reported patients with SGPL1 mutations and compared them to the presented patients. RESULTS The median age at kidney presentation was 5 months. Four patients (67%) were diagnosed before age 1 year. Kidney biopsy showed focal segmental glomerulosclerosis in 2 patients and diffuse mesangial sclerosis in one patient. Steroids were given to 3 patients, but they did not respond. All 6 patients progressed to chronic kidney disease; 5 required kidney replacement therapy (KRT) at a median age of 6 months. Deceased kidney transplantation was performed in one patient. All 6 patients had adrenal insufficiency, of which 5 were diagnosed at age < 6 months. Three patients had hypothyroidism, 2 had ichthyosis, 4 had immunodeficiency, 5 had neurological findings, and 2 had genitourinary system anomalies. Four patients died at a median age of 30.5 months. Two patients are being followed up with KRT. One patient had a novel mutation. CONCLUSIONS Patients with SGPL1 mutations have a poor prognosis, and many types of extrarenal organ/system involvement beyond adrenal insufficiency can be seen. Genetic diagnosis of such patients is important for treatment, genetic counseling, and screening for comorbid conditions. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Tugba Tastemel Ozturk
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey.
| | - Nur Canpolat
- Department of Pediatric Nephrology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Seha Saygili
- Department of Pediatric Nephrology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Umut Selda Bayrakci
- Department of Pediatric Nephrology, Faculty of Medicine, Ankara City Hospital, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Oguz Soylemezoglu
- Department of Pediatric Nephrology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Fatih Ozaltin
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
- Nephrogenetics Laboratory, Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, Ankara, Turkey
| | - Rezan Topaloglu
- Department of Pediatric Nephrology, Faculty of Medicine, Hacettepe University, Sihhiye, 06100, Ankara, Turkey
| |
Collapse
|
35
|
Hureaux M, Heidet L, Vargas-Poussou R, Dorval G. [Major advances in pediatric nephro-genetics]. Med Sci (Paris) 2023; 39:234-245. [PMID: 36943120 DOI: 10.1051/medsci/2023028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2023] Open
Abstract
The rise of genetics in the last decades has allowed major advances in the understanding of the mechanisms leading to inherited kidney diseases. From the first positional cloning studies to the advent of high-throughput sequencing (NGS), genome analysis technologies have become increasingly efficient, with an extraordinary level of resolution. Moreover, sequencing prices have decreased from one million dollars for the sequencing of James Watson's genome in 2008, to a few hundred dollars for the sequencing of a genome today. Thus, molecular diagnosis has a central place in the diagnosis of these patients and influences the therapeutic management in many situations. However, although NGS is a powerful tool for the identification of variants involved in diseases, it also exposes to the risk of over-interpretation of certain variants, leading to erroneous diagnoses, requiring the use of specialists. In this review, we first propose a brief retrospective of the essential steps that led to the current knowledge and the development of NGS for the study of hereditary nephropathies in children. This review is then an opportunity to present the main hereditary nephropathies and the underlying molecular mechanisms. Among them, we emphasize ciliopathies, congenital anomalies of the kidney and urinary tract, podocytopathies and tubulopathies.
Collapse
Affiliation(s)
- Marguerite Hureaux
- Service de médecine génomique des maladies rares, AP-HP, université Paris Cité, France - Inserm U970, Paris CardioVascular Research Center, université Paris Cité, faculté de médecine, France - Centre de référence des maladies rénales héréditaires de l'enfant et de l'adulte MARHEA, hôpital Necker-Enfants Malades, Paris, France
| | - Laurence Heidet
- Centre de référence des maladies rénales héréditaires de l'enfant et de l'adulte MARHEA, hôpital Necker-Enfants Malades, Paris, France - Service de néphrologie pédiatrique, AP-HP, université Paris Cité, France - CNRS, centre de recherche des Cordeliers, Inserm UMRS 1138, Sorbonne université, université Paris Cité, France
| | - Rosa Vargas-Poussou
- Service de médecine génomique des maladies rares, AP-HP, université Paris Cité, France - Centre de référence des maladies rénales héréditaires de l'enfant et de l'adulte MARHEA, hôpital Necker-Enfants Malades, Paris, France - CNRS, centre de recherche des Cordeliers, Inserm UMRS 1138, Sorbonne université, université Paris Cité, France
| | - Guillaume Dorval
- Service de médecine génomique des maladies rares, AP-HP, université Paris Cité, France - Centre de référence des maladies rénales héréditaires de l'enfant et de l'adulte MARHEA, hôpital Necker-Enfants Malades, Paris, France - Inserm U1163, Laboratoire des maladies rénales héréditaires, institut Imagine, université Paris Cité, France
| |
Collapse
|
36
|
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
|
37
|
Hendriks WJAJ, van Cruchten RTP, Pulido R. Hereditable variants of classical protein tyrosine phosphatase genes: Will they prove innocent or guilty? Front Cell Dev Biol 2023; 10:1051311. [PMID: 36755664 PMCID: PMC9900141 DOI: 10.3389/fcell.2022.1051311] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
Protein tyrosine phosphatases, together with protein tyrosine kinases, control many molecular signaling steps that control life at cellular and organismal levels. Impairing alterations in the genes encoding the involved proteins is expected to profoundly affect the quality of life-if compatible with life at all. Here, we review the current knowledge on the effects of germline variants that have been reported for genes encoding a subset of the protein tyrosine phosphatase superfamily; that of the thirty seven classical members. The conclusion must be that the newest genome research tools produced an avalanche of data that suggest 'guilt by association' for individual genes to specific disorders. Future research should face the challenge to investigate these accusations thoroughly and convincingly, to reach a mature genotype-phenotype map for this intriguing protein family.
Collapse
Affiliation(s)
- Wiljan J. A. J. Hendriks
- Department of Cell Biology, Radboud University Medical Centre, Nijmegen, The Netherlands,*Correspondence: Wiljan J. A. J. Hendriks,
| | | | - Rafael Pulido
- Biomarkers in Cancer Unit, Biocruces Bizkaia Health Research Institute, Barakaldo, Spain,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
38
|
Abdou M, Ramadan A, El-Agamy BE, EL-Farsy MS, Saleh EM. Mutational analysis of phospholipase C epsilon 1 gene in Egyptian children with steroid-resistant nephrotic syndrome. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2022. [DOI: 10.1186/s43042-022-00353-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Steroid-resistant nephrotic syndrome (SRNS) is characterized by unresponsiveness of nephrotic range proteinuria to standard steroid therapy, and is the main cause of childhood renal failure. The identification of more than 53 monogenic causes of SRNS has led researchers to focus on the genetic mutations related to the molecular mechanisms of the disease. Mutations in the PLCE1 gene, which encodes phospholipase C epsilon 1 (PLCε1), have been described in patients with early-onset SRNS characterized by progressive renal failure. In this study we screened for PLCE1 mutations in Egyptian children with SRNS. This is a descriptive case series study aiming to screen for PLCE1 gene mutations by direct sequencing of five exons—9, 12, 15, 19, 27—in 20 Egyptian children with SRNS who entered the Nephrology Unit, Faculty of Medicine, Ain-Shams University from November 2015 to December 2017. The variants detected were submitted to in silico analysis.
Results
We screened for mutations in five selected exons of PLCE1 gene. We identified seven variants in the five selected exons with homozygous and heterozygous inheritance pattern, two are intronic variants, two are silent variants, and three are missense variants. We identified four novel variants two are silent with no clinical significance and two are missense with uncertain clinical significance and pathogenic in-silico predictions; one p.Arg1230His in exon 12, the other is p.Glu1393Lys in exon 15.
Conclusions
We identified four novel mutations, findings which added to the registered SNP spectrum of the PLCE1 gene. These results widen the spectrum of PLCE1 gene mutations and support the importance of genetic testing in different populations of SRNS patients, therefore, to assess the vulnerability of Egyptian children to SRNS candidate genes, further studies needed on a larger number of cases which undoubtedly provide new insights into the pathogenic mechanisms of SRNS and might help in control of the patient. Additionally, the use of computational scoring and modeling tools may assist in the evaluation of the way in which the SNPs affect protein functionality.
Collapse
|
39
|
Mishra OP, Sidar M, Batra VV, Prasad R, Singh A, Abhinay A, Mishra A, Yadav AK. Desfechos de crianças com síndrome nefrótica idiopática córtico-resistente: um estudo observacional de centro único. J Bras Nefrol 2022. [DOI: 10.1590/2175-8239-jbn-2022-0073pt] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RESUMO Introdução: A síndrome nefrótica idiopática córtico-resistente (SNICR) apresenta desfechos variáveis em crianças. O objetivo principal deste estudo foi avaliar a taxa de remissão cumulativa. Os objetivos secundários foram avaliar fatores que afetam status de remissão, sobrevida da função renal e efeitos adversos de medicamentos. Métodos: Foram incluídos 114 pacientes com SNCR. Utilizou-se protocolo de tratamento baseado em inibidores de calcineurina juntamente com prednisolona e inibidor da enzima conversora de angiotensina. Os pacientes foram acompanhados durante 5 anos. Resultados: A idade mediana foi 4,5 anos; 53,5% dos casos tinham entre 1 e 5 anos. 62 pacientes (54,4%) estavam em estágio inicial; 52 (45,6%) em estágio tardio da SNCR. A TFGecr mediana foi 83,5 mL/min/1,73 m2 na apresentação. Dos 110 pacientes, 63 (57,3%) alcançaram remissão [remissão completa 30 (27,3%), remissão parcial 33 (30%)], e 47 (42,7%) não apresentaram remissão. A sobrevida da função renal foi 87,3%; 14 casos (12,7%) progrediram para DRC (G3-8, G4-3, G5-1, G5D-2). A duração mediana do acompanhamento foi 36 meses (IIQ 24, 60). Idade no início, ciclosporina/tacrolimus, TFGecr e histopatologia (DLM/GESF) não afetaram a remissão. Igualmente, status de remissão, além da idade no início, protocolo de medicamentos e histopatologia não afetaram significativamente a função renal por 5 anos. Observou-se hipertensão, fácies cushingoide, baixa estatura, catarata e obesidade em 37,7; 29,8; 25,5; 17,5; e 0,7% dos casos, respectivamente. Conclusão: Aproximadamente metade dos casos alcançou remissão. Idade no início, uso de ciclosporina/tacrolimus e lesão histopatológica não afetaram o status de remissão nem a sobrevida da função renal a curto prazo na SNICR.
Collapse
Affiliation(s)
| | | | - Vineeta V. Batra
- G. B. Pant Institute of Post Graduate Medical Education & Research, India
| | | | | | | | - Akash Mishra
- Jawahar Lal Institute of Postgraduate Medical Education and Research, India
| | | |
Collapse
|
40
|
Ilori T, Watanabe A, Ng KH, Solarin A, Sinha A, Gbadegesin R. Genetics of Chronic Kidney Disease in Low-Resource Settings. Semin Nephrol 2022; 42:151314. [PMID: 36801667 PMCID: PMC10272019 DOI: 10.1016/j.semnephrol.2023.151314] [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] [Indexed: 02/19/2023]
Abstract
Advances in kidney genomics in the past 20 years has opened the door for more precise diagnosis of kidney disease and identification of new and specific therapeutic agents. Despite these advances, an imbalance exists between low-resource and affluent regions of the world. Individuals of European ancestry from the United States, United Kingdom, and Iceland account for 16% of the world's population, but represent more than 80% of all genome-wide association studies. South Asia, Southeast Asia, Latin America, and Africa together account for 57% of the world population but less than 5% of genome-wide association studies. Implications of this difference include limitations in new variant discovery, inaccurate interpretation of the effect of genetic variants in non-European populations, and unequal access to genomic testing and novel therapies in resource-poor regions. It also further introduces ethical, legal, and social pitfalls, and ultimately may propagate global health inequities. Ongoing efforts to reduce the imbalance in low-resource regions include funding and capacity building, population-based genome sequencing, population-based genome registries, and genetic research networks. More funding, training, and capacity building for infrastructure and expertise is needed in resource-poor regions. Focusing on this will ensure multiple-fold returns on investments in genomic research and technology.
Collapse
Affiliation(s)
- Titilayo Ilori
- Division of Nephrology, Boston University School of Medicine, Boston, MA
| | - Andreia Watanabe
- Division of Molecular Medicine, Department of Pediatrics, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Kar-Hui Ng
- Department of Pediatrics, Yong Loo Lin School of Medicine, Singapore
| | - Adaobi Solarin
- Department of Pediatrics and Child Health, Lagos State University College of Medicine, Ikeja, Lagos, Nigeria
| | - Aditi Sinha
- Department of Pediatrics, All India Institute of Medical Sciences, New Delhi, India
| | - Rasheed Gbadegesin
- Division of Nephrology, Department of Pediatrics, Duke University School of Medicine, Durham, NC.
| |
Collapse
|
41
|
Sinha R, Pradhan S, Banerjee S, Jahan A, Akhtar S, Pahari A, Raut S, Parakh P, Basu S, Srivastava P, Nayak S, Thenral SG, Ramprasad V, Ashton E, Bockenhauer D, Mandal K. Whole-exome sequencing and variant spectrum in children with suspected inherited renal tubular disorder: the East India Tubulopathy Gene Study. Pediatr Nephrol 2022; 37:1811-1836. [PMID: 35006361 DOI: 10.1007/s00467-021-05388-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/29/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Inherited tubulopathies are a heterogeneous group of genetic disorders making whole-exome sequencing (WES) the preferred diagnostic methodology. METHODS This was a multicenter descriptive study wherein children (< 18 years) with clinically suspected tubular disorders were recruited for molecular testing through WES. Multiplex ligation-dependent probe amplification (MLPA) and Sanger sequencing were done when required. Variants were classified as per American College of Medical Genetics 2015 guidelines and pathogenic (P)/likely pathogenic (LP) variants were considered causative. RESULTS There were 77 index cases (male =73%). Median age at diagnosis was 48 months (IQR 18.5 to 108 months). At recruitment, the number of children in each clinical group was as follows: distal renal tubular acidosis (dRTA) = 25; Bartter syndrome = 18; isolated hypophosphatemic rickets (HP) = 6; proximal tubular dysfunction (pTD) = 12; nephrogenic diabetes insipidus (NDI) = 6; kidney stone/nephrocalcinosis (NC) = 6; others = 4. We detected 55 (24 novel) P/LP variants, providing genetic diagnoses in 54 children (70%). The diagnostic yield of WES was highest for NDI (100%), followed by HP (83%; all X-linked HP), Bartter syndrome (78%), pTD (75%), dRTA (64%), and NC (33%). Molecular testing had a definite impact on clinical management in 24 (31%) children. This included revising clinical diagnosis among 14 children (26% of those with a confirmed genetic diagnosis and 18% of the overall cohort), detection of previously unrecognized co-morbidities among 8 children (sensorineural deafness n = 5, hemolytic anemia n = 2, and dental changes n = 1) and facilitating specific medical treatment for 7 children (primary hyperoxaluria n = 1, cystinosis n = 4, tyrosinemia n = 2). CONCLUSION WES is a powerful tool in the diagnosis and management of children with inherited tubulopathies in the Indian population. A higher resolution version of the Graphical abstract is available as Supplementary information.
Collapse
Affiliation(s)
- Rajiv Sinha
- Institute of Child Health, Kolkata, India
- Apollo Hospital, Kolkata, India
| | | | - Sushmita Banerjee
- Institute of Child Health, Kolkata, India
- Calcutta Medical and Research Institute, Kolkata, India
| | - Afsana Jahan
- Renowell Clinic and Pratiksha Hospital, Gauhati, India
| | | | | | | | | | | | - Priyanka Srivastava
- Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | | | | | - Emma Ashton
- Rare & Inherited Disease Laboratory, NHS North Thames Genomic Laboratory Hub, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Detlef Bockenhauer
- UCL Department of Renal Medicine and Renal Unit, Great Ormond Street Hospital for Children, NHS Foundation Trust, London, UK
| | - Kausik Mandal
- Department of Medical Genetics, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, 226014, India.
| |
Collapse
|
42
|
Dirim AB, Demir E, Guller N, Safak S, Artan AS, Oto OA, Ozluk Y, Ozturk S, Yazici H, Kalayoglu-Besisik S, Turkmen A. Efficacy of intravenous combined immunosuppression with plasmapheresis in adult patients with refractory primary focal segmental glomerulosclerosis. J Clin Apher 2022; 37:376-387. [PMID: 35535432 DOI: 10.1002/jca.21985] [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: 12/08/2021] [Revised: 04/17/2022] [Accepted: 04/18/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Primary focal segmental glomerulosclerosis (FSGS) treatment is based on immunosuppressive therapies. Since refractory disease is common, alternative methods are emerging. One of these methods is plasmapheresis with intravenous cyclosporine and corticosteroids, and it could be an option in post-transplant recurrent FSGS. We retrospectively investigated the efficacy of this combined treatment in adult patients with refractory primary FSGS. METHODS Seven refractory primary FSGS patients were included. Demographics, estimated glomerular filtration rates, serum albumin levels, urine protein/creatinine ratios, and previous treatments were evaluated. Also, complications and remission rates were assessed. RESULTS Median patient age was 23 years. Median duration of diagnosis was 2 years. Median number of plasmapheresis sessions was 14. Five of seven patients (71.4%, one complete, four partial remissions) were responders after the protocol. Changes in serum albumin levels and proteinuria after protocol were statistically significant (P = 0.018 and P = 0.018, respectively). eGFR levels did not change statistically (P = 0.753). Median follow-up duration after the treatment was 17 months. However, two patients experienced disease relapse (28.5%). End-stage kidney disease was developed in two patients. Sustained remission rate was 42.8% during follow-up (One complete and two partial remissions). Also, 42.8% of patients experienced catheter infections. Catheter-associated thrombosis that required surgery was observed in a patient. CONCLUSIONS Plasmapheresis combined with intravenous cyclosporine and corticosteroids could be an option in refractory primary FSGS. High response rates after this protocol were encouraging. However, the relapsing disease was observed after the cessation of apheresis. Also, complications of the protocol could limit the applicability.
Collapse
Affiliation(s)
- Ahmet Burak Dirim
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Erol Demir
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nurane Guller
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Seda Safak
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ayse Serra Artan
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Ozgur Akin Oto
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Yasemin Ozluk
- Department of Pathology, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Savas Ozturk
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Halil Yazici
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Sevgi Kalayoglu-Besisik
- Division of Hematology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Aydin Turkmen
- Division of Nephrology, Department of Internal Medicine, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| |
Collapse
|
43
|
Lu L, Yap YC, Nguyen DQ, Chan YH, Ng JL, Zhang YC, Chan CY, Than M, Liu ID, Asim S, Moorani K, Naeem B, Ijaz I, Nguyen TMT, Lee ML, Eng C, Huque SS, Ng YH, Ganesan I, Chao SM, Chong SL, Tan PH, Loh A, Davila S, Kumar V, Ling JZJ, Moorakonda RB, Tan KML, Ng AYJ, Poon KS, Schaefer F, Lipska-Zietkiewicz B, Yap HK, Ng KH. Multicenter study on the genetics of glomerular diseases among southeast and south Asians: Deciphering Diversities - Renal Asian Genetics Network (DRAGoN). Clin Genet 2022; 101:541-551. [PMID: 35064937 DOI: 10.1111/cge.14116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 11/30/2022]
Abstract
Multinational studies have reported monogenic etiologies in 25%-30% of children with steroid-resistant nephrotic syndrome. Such large studies are lacking in Asia. We established Deciphering Diversities: Renal Asian Genetics Network (DRAGoN) and aimed to describe the genetic and clinical spectrums in Asians. We prospectively studied a cohort of 183 probands with suspected genetic glomerulopathies from South and Southeast Asia, of whom 17% had positive family history. Using multi-gene panel sequencing, we detected pathogenic variants in 26 (14%) probands, of whom one-third had COL4A4 or COL4A5 variants (n = 9, 5%). Of those with COL4A5 defects, only 25% had features suggestive of Alport syndrome. Besides traditional predictors for genetic disease (positive family history and extrarenal malformations), we identified novel predictors, namely older age (6.2 vs. 2.4 years; p = 0.001), hematuria (OR 5.6; 95% CI 2.1-14.8; p < 0.001), and proteinuria in the absence of nephrotic syndrome (OR 4.6; 95% CI 1.8-11.8; p = 0.001) at first manifestation. Among patients who first presented with proteinuria without nephrotic syndrome, the genetic diagnostic rates were >60% when a second risk factor (positive family history or extrarenal manifestation) co-existed. The genetic spectrum of glomerulopathies appears different in Asia. Collagen IV genes may be included in sequencing panels even when suggestive clinical features are absent.
Collapse
Affiliation(s)
- Liangjian Lu
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Yok-Chin Yap
- Department of Paediatrics, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Duc Quang Nguyen
- Nephrology and Endocrinology Department, Children's Hospital 1, Ho Chi Minh, Vietnam
| | - Yiong-Huak Chan
- Biostatistics, National University of Singapore, Singapore, Singapore
| | - Jun-Li Ng
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
- Paediatrics, National University of Singapore, Singapore, Singapore
| | - Yao-Chun Zhang
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
- Paediatrics, National University of Singapore, Singapore, Singapore
| | - Chang-Yien Chan
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
- Paediatrics, National University of Singapore, Singapore, Singapore
| | - Mya Than
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
- Paediatrics, National University of Singapore, Singapore, Singapore
| | - Isaac Desheng Liu
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Sadaf Asim
- Paediatric Nephrology, National Institute of Child Health, Karachi, Pakistan
| | - Khemchand Moorani
- Paediatric Nephrology, National Institute of Child Health, Karachi, Pakistan
| | - Bilquis Naeem
- Paediatric Nephrology, National Institute of Child Health, Karachi, Pakistan
| | - Iftikhar Ijaz
- Department of Pediatrics, King Edward Medical University, Lahore, Pakistan
| | | | - Ming-Lee Lee
- Department of Paediatrics, Hospital Tuanku Ja'afar Seremban, Seremban, Malaysia
| | - Caroline Eng
- Department of Paediatrics, Hospital Tuanku Ja'afar Seremban, Seremban, Malaysia
| | - Syed Saimul Huque
- Paediatric Nephrology, Bangabandhu Sheikh Mujib Medical University, Dhaka, Bangladesh
| | - Yong-Hong Ng
- Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Indra Ganesan
- Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Sing-Ming Chao
- Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Siew-Le Chong
- Paediatrics, KK Women's and Children's Hospital, Singapore, Singapore
| | - Puay-Hoon Tan
- Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Alwin Loh
- Anatomical Pathology, Singapore General Hospital, Singapore, Singapore
| | - Sonia Davila
- Institute of Precision Medicine, Duke-NUS, Singapore, Singapore
| | - Vikrant Kumar
- Cancer and Stem Cell Biology, Duke-NUS, Singapore, Singapore
| | | | | | - Karen Mei-Ling Tan
- Molecular Diagnosis Centre, National University Hospital, Singapore, Singapore
| | - Alvin Yu-Jin Ng
- Molecular Diagnosis Centre, National University Hospital, Singapore, Singapore
| | - Kok-Siong Poon
- Molecular Diagnosis Centre, National University Hospital, Singapore, Singapore
| | - Franz Schaefer
- Department of Paediatrics Nephrology, Center for Paediatrics and Adolescent Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Hui-Kim Yap
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
- Paediatrics, National University of Singapore, Singapore, Singapore
| | - Kar-Hui Ng
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
- Paediatrics, National University of Singapore, Singapore, Singapore
| |
Collapse
|
44
|
Massengill S, Trachtman H. Genetic Spectrum of Nephrotic Syndrome: Impact of Podocytopathy in Adult Life. Adv Chronic Kidney Dis 2022; 29:221-224. [PMID: 36084968 DOI: 10.1053/j.ackd.2022.04.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/11/2022] [Accepted: 04/12/2022] [Indexed: 11/11/2022]
Abstract
A substantial number of patients with focal segmental glomerulosclerosis (FSGS) have a pathogenic genetic mutation in a podocyte protein as the cause of their disease. The mutations can affect a wide range of cell functions including the actin cytoskeleton, cell adhesion and motility, mitochondrial function, and nuclear pore proteins. The likelihood of a genetic cause declines with age, from approximately 30% in children and adolescents to 10% in adulthood, and the specific proteins involved and the pattern of inheritance differ in the 2 age groups. The presence of a genetic cause for FSGS can have important clinical ramifications including the need for a diagnostic kidney biopsy, medical management, and the risk of recurrent disease after kidney transplantation. This review summarizes the spectrum of genetic causes of nephrotic syndrome, primarily FSGS, in adults with a focus on diagnosis, presentation, and management.
Collapse
Affiliation(s)
| | - Howard Trachtman
- Department of Pediatrics, University of Michigan, Ann Arbor, MI.
| |
Collapse
|
45
|
Jiao J, Wang L, Ni F, Wang M, Feng S, Gao X, Chan H, Yang X, Lee H, Chi H, Chen X, Wu D, Zhang G, Yang B, Wang A, Yang Q, Wan J, Yu S, Li X, Wang M, Chen X, Mai X, Ruan X, Yang H, Li Q. Whole-exome sequencing of a multicenter cohort identifies genetic changes associated with clinical phenotypes in pediatric nephrotic syndrome. Genes Dis 2022; 9:1662-1673. [PMID: 36157477 PMCID: PMC9485284 DOI: 10.1016/j.gendis.2022.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 03/06/2022] [Accepted: 03/22/2022] [Indexed: 11/19/2022] Open
Abstract
Understanding the association between the genetic and clinical phenotypes in children with nephrotic syndrome (NS) of different etiologies is critical for early clinical guidance. We employed whole-exome sequencing (WES) to detect monogenic causes of NS in a multicenter cohort of 637 patients. In this study, a genetic cause was identified in 30.0% of the idiopathic steroid-resistant nephrotic syndrome (SRNS) patients. Other than congenital nephrotic syndrome (CNS), there were no significant differences in the incidence of monogenic diseases based on the age at manifestation. Causative mutations were detected in 39.5% of patients with focal segmental glomerulosclerosis (FSGS) and 9.2% of those with minimal change disease (MCD). In terms of the patterns in patients with different types of steroid resistance, a single gene mutation was identified in 34.8% of patients with primary resistance, 2.9% with secondary resistance, and 71.4% of children with multidrug resistance. Among the various intensified immunosuppressive therapies, tacrolimus (TAC) showed the highest response rate, with 49.7% of idiopathic SRNS patients achieving complete remission. Idiopathic SRNS patients with monogenic disease showed a similar multidrug resistance pattern, and only 31.4% of patients with monogenic disease achieved a partial remission on TAC. During an average 4.1-year follow-up, 21.4% of idiopathic SRNS patients with monogenic disease progressed to end-stage renal disease (ESRD). Collectively, this study provides evidence that genetic testing is necessary for presumed steroid-resistant and idiopathic SRNS patients, especially those with primary and/or multidrug resistance.
Collapse
Affiliation(s)
- Jia Jiao
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Li Wang
- Department of Nephrology, Chengdu Women and Children Central Hospital, Chengdu, Sichuan 610091, PR China
| | - Fenfen Ni
- Department of Nephrology, Sheen Children's Hospital, Shenzhen, Guangdong 518034, PR China
| | - Mo Wang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Shipin Feng
- Department of Nephrology, Chengdu Women and Children Central Hospital, Chengdu, Sichuan 610091, PR China
| | - Xiaojie Gao
- Department of Nephrology, Sheen Children's Hospital, Shenzhen, Guangdong 518034, PR China
| | - Han Chan
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Xueying Yang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Hao Lee
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Huan Chi
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Xuelan Chen
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Daoqi Wu
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Gaofu Zhang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Baohui Yang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Anshuo Wang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Qin Yang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Junli Wan
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Sijie Yu
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Xiaoqin Li
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Mei Wang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Xiaofeng Chen
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Xianying Mai
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
| | - Xiongzhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, PR China
- John Moorhead Research Laboratory, Centre for Nephrology, University College London Medical School, Royal Free Campus, University College London, London WC1E 6BT, United Kingdom
| | - Haiping Yang
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
- Corresponding author.
| | - Qiu Li
- Department of Nephrology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400015, PR China
- Corresponding author.
| |
Collapse
|
46
|
Drovandi S, Lipska-Ziętkiewicz BS, Ozaltin F, Emma F, Gulhan B, Boyer O, Trautmann A, Xu H, Shen Q, Rao J, Riedhammer KM, Heemann U, Hoefele J, Stenton SL, Tsygin AN, Ng KH, Fomina S, Benetti E, Aurelle M, Prikhodina L, Schreuder MF, Tabatabaeifar M, Jankowski M, Baiko S, Mao J, Feng C, Liu C, Sun S, Deng F, Wang X, Clavé S, Stańczyk M, Bałasz-Chmielewska I, Fila M, Durkan AM, Levart TK, Dursun I, Esfandiar N, Haas D, Bjerre A, Anarat A, Benz MR, Talebi S, Hooman N, Ariceta G, Schaefer F. Oral Coenzyme Q10 supplementation leads to better preservation of kidney function in steroid resistant nephrotic syndrome due to primary Coenzyme Q10 deficiency. Kidney Int 2022; 102:604-612. [DOI: 10.1016/j.kint.2022.04.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/13/2022] [Accepted: 04/29/2022] [Indexed: 12/17/2022]
|
47
|
Leenen E, Erger F, Altmüller J, Wenzel A, Thiele H, Harth A, Tschernoster N, Lokhande S, Joerres A, Becker JU, Ekici A, Huettel B, Beck B, Weidemann A. Alport syndrome and autosomal dominant tubulointerstitial kidney disease frequently underlie end stage renal disease of unknown origin - a single center analysis. Nephrol Dial Transplant 2022; 37:1895-1905. [PMID: 35485766 DOI: 10.1093/ndt/gfac163] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The prevalence of end stage renal disease of unknown etiology in adult patients is globally high and accounts for almost 20% of all dialysis patients. Recent studies have suggested that the percentage of adult patients with a causal genetic variant has been underestimated so far. Despite severe prognostic and therapeutic implications, awareness about prevalence and manifestations of genetic kidney diseases in adult renal patients is still limited. MATERIALS AND METHODS We recruited 58 individuals from 39 families at our transplantation center, fulfilling at least one of the following criteria: 1) unclear etiology of kidney disease 2) clinically suspected genetic kidney disease 3) positive family history for nephropathies. The cohort consisted of patients waitlisted for kidney transplantation and patients in the follow-up after transplantation. Detailed documentation of family history and phenotype was obtained before initiating gene panel sequencing of 479 nephropathy-associated genes. RESULTS With this study design, a molecular genetic diagnosis was established in one third of all patients. Mutations in the collagen COL4A-genes, and mutations in MUC1 and UMOD were the most frequent among all detected causal variants. Overall, rare genetic variants were detected in more than half of all cases. CONCLUSION The combination of detailed phenotyping prior to NGS diagnostics was highly efficient. Elucidating the underlying genetic causes in a cohort of adult renal patients has considerable clinical impact on medical management.
Collapse
Affiliation(s)
- Esther Leenen
- Department of Medicine I - Nephrology, Transplantation & Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Germany
| | - Florian Erger
- Institute of Human Genetics, Center for Molecular Medicine Cologne and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Janine Altmüller
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Core Facility Genomics, Berlin, Germany.,Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Andrea Wenzel
- Institute of Human Genetics, Center for Molecular Medicine Cologne and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Holger Thiele
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Ana Harth
- Department of Medicine I - Nephrology, Transplantation & Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Germany
| | - Nikolai Tschernoster
- Institute of Human Genetics, Center for Molecular Medicine Cologne and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany.,Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | - Shanti Lokhande
- Department of Medicine I - Nephrology, Transplantation & Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Germany
| | - Achim Joerres
- Department of Medicine I - Nephrology, Transplantation & Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Germany
| | - Jan-Ulrich Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Arif Ekici
- Institute of Human Genetics, University Hospital Erlangen, Germany
| | - Bruno Huettel
- Max-Plank-Genome-Centre Cologne (MP-GC), Cologne, Germany
| | - Bodo Beck
- Institute of Human Genetics, Center for Molecular Medicine Cologne and Center for Rare and Hereditary Kidney Disease, Cologne, University Hospital of Cologne, Cologne, Germany
| | - Alexander Weidemann
- Department of Medicine I - Nephrology, Transplantation & Medical Intensive Care, University Witten/Herdecke, Medical Center Cologne-Merheim, Germany.,Department of Nephrology, St. Vincenz Hospital, Paderborn, Germany
| |
Collapse
|
48
|
Drovandi S, Lipska-Ziętkiewicz BS, Ozaltin F, Emma F, Gulhan B, Boyer O, Trautmann A, Ziętkiewicz S, Xu H, Shen Q, Rao J, Riedhammer KM, Heemann U, Hoefele J, Stenton SL, Tsygin AN, Ng KH, Fomina S, Benetti E, Aurelle M, Prikhodina L, Schijvens AM, Tabatabaeifar M, Jankowski M, Baiko S, Mao J, Feng C, Deng F, Rousset-Rouviere C, Stańczyk M, Bałasz-Chmielewska I, Fila M, Durkan AM, Levart TK, Dursun I, Esfandiar N, Haas D, Bjerre A, Anarat A, Benz MR, Talebi S, Hooman N, Ariceta G, Schaefer F. Variation of the clinical spectrum and genotype-phenotype associations in Coenzyme Q10 deficiency associated glomerulopathy. Kidney Int 2022; 102:592-603. [PMID: 35483523 DOI: 10.1016/j.kint.2022.02.040] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/21/2022] [Accepted: 02/28/2022] [Indexed: 12/14/2022]
Abstract
Primary Coenzyme Q10 deficiency is a rare mitochondriopathy with a wide spectrum of organ involvement, including steroid-resistant nephrotic syndrome mainly associated with disease-causing variants in the genes COQ2, COQ6 or COQ8B. We performed a systematic literature review, PodoNet, MitoNET,and CCGKDD registries queries and an online survey, collecting comprehensive clinical and genetic data of 251 patients spanning 173 published (47 updated) and 78 new cases. Kidney disease was first diagnosed at median age 1.0, 1.2 and 9.8 years in individuals with disease-causing variants in COQ2, COQ6 and COQ8B, respectively. Isolated kidney involvement at diagnosis occurred in 34% of COQ2, 10.8% of COQ6 and 70.7% of COQ8B variant individuals. Classic infantile multiorgan involvement comprised 22% of the COQ2 variant cohort while 47% of them developed neurological symptoms at median age 2.7 years. The association of steroid-resistant nephrotic syndrome and sensorineural hearing loss was confirmed as the distinctive phenotype of COQ6 variants, with hearing impairment manifesting at average age three years. None of the patients with COQ8B variants, but 50% of patients with COQ2 and COQ6 variants progressed to kidney failure by age five. At adult age, kidney survival was equally poor (20-25%) across all disorders. A number of sequence variants, including putative local founder mutations, had divergent clinical presentations, in terms of onset age, kidney and non-kidney manifestations and kidney survival. Milder kidney phenotype was present in those with biallelic truncating variants within the COQ8B variant cohort. Thus, significant intra- and inter-familial phenotype variability was observed, suggesting both genetic and non-genetic modifiers of disease severity.
Collapse
|
49
|
Devarajan P, Chertow GM, Susztak K, Levin A, Agarwal R, Stenvinkel P, Chapman AB, Warady BA. Emerging Role of Clinical Genetics in CKD. Kidney Med 2022; 4:100435. [PMID: 35372818 PMCID: PMC8971313 DOI: 10.1016/j.xkme.2022.100435] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Chronic kidney disease (CKD) afflicts 15% of adults in the United States, of whom 25% have a family history. Genetic testing is supportive in identifying and possibly confirming diagnoses of CKD, thereby guiding care. Advances in the clinical genetic evaluation include next-generation sequencing with targeted gene panels, whole exome sequencing, and whole genome sequencing. These platforms provide DNA sequence reads with excellent coverage throughout the genome and have identified novel genetic causes of CKD. New pathologic genetic variants identified in previously unrecognized biological pathways have elucidated disease mechanisms underlying CKD etiologies, potentially establishing prognosis and guiding treatment selection. Molecular diagnoses using genetic sequencing can detect rare, potentially treatable mutations, avoid misdiagnoses, guide selection of optimal therapy, and decrease the risk of unnecessary and potentially harmful interventions. Genetic testing has been widely adopted in pediatric nephrology; however, it is less frequently used to date in adult nephrology. Extension of clinical genetic approaches to adult patients may achieve similar benefits in diagnostic refinement and treatment selection. This review aimed to identify clinical CKD phenotypes that may benefit the most from genetic testing, outline the commonly available platforms, and provide examples of successful deployment of these approaches in CKD.
Collapse
Affiliation(s)
- Prasad Devarajan
- Division of Nephrology and Hypertension, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, OH
| | | | - Katalin Susztak
- Division of Nephrology, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Adeera Levin
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rajiv Agarwal
- Division of Nephrology, Indiana University, Indianapolis, IN
| | - Peter Stenvinkel
- Department of Renal Medicine, Karolinska University Hospital at Huddinge, Karolinkska Institutet, Stockholm, Sweden
| | | | - Bradley A. Warady
- Division of Pediatric Nephrology, Children’s Mercy Kansas City, Kansas City, MO
| |
Collapse
|
50
|
Sinha R, Chaudhury AR, Sarkar S, Banerjee S, Pulai S, Dasgupta S, Muorah M, Datta D. High incidence of COL4Ageneticvariants among a cohort of childhood with steroid resistance nephrotic syndrome from Eastern India. Kidney Int Rep 2022; 7:913-915. [PMID: 35497790 PMCID: PMC9039466 DOI: 10.1016/j.ekir.2022.01.1047] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/06/2022] [Accepted: 01/10/2022] [Indexed: 11/25/2022] Open
|