51
|
Milone R, Tancredi R, Cosenza A, Ferrari AR, Scalise R, Cioni G, Battini R. 17q12 Recurrent Deletions and Duplications: Description of a Case Series with Neuropsychiatric Phenotype. Genes (Basel) 2021; 12:genes12111660. [PMID: 34828266 PMCID: PMC8620923 DOI: 10.3390/genes12111660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/22/2022] Open
Abstract
Syndromic neurodevelopmental disorders are usually investigated through genetics technologies, within which array comparative genomic hybridization (Array-CGH) is still considered the first-tier clinical diagnostic test. Among recurrent syndromic imbalances, 17q12 deletions and duplications are characterized by neurodevelopmental disorders associated with visceral developmental disorders, although expressive variability is common. Here we describe a case series of 12 patients with 17q12 chromosomal imbalances, in order to expand the phenotypic characterization of these recurrent syndromes whose diagnosis is often underestimated, especially if only mild traits are present. Gene content and genotype-phenotype correlations have been discussed, with special regard to neuropsychiatric features, whose impact often requires etiologic analysis.
Collapse
Affiliation(s)
- Roberta Milone
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
| | - Raffaella Tancredi
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
| | - Angela Cosenza
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
| | - Anna Rita Ferrari
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
| | - Roberta Scalise
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
- Tuscan PhD Program of Neuroscience, University of Florence, Pisa and Siena, 50139 Florence, Italy
| | - Giovanni Cioni
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Fondazione Stella Maris, Calambrone, 56128 Pisa, Italy; (R.M.); (R.T.); (A.C.); (A.R.F.); (R.S.); (G.C.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
- Correspondence:
| |
Collapse
|
52
|
Piedrafita A, Balayssac S, Casemayou A, Saulnier-Blache JS, Lucas A, Iacovoni JS, Breuil B, Chauveau D, Decramer S, Malet-Martino M, Schanstra JP, Faguer S. Hepatocyte nuclear factor-1β shapes the energetic homeostasis of kidney tubule cells. FASEB J 2021; 35:e21931. [PMID: 34653285 DOI: 10.1096/fj.202100782rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/28/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022]
Abstract
Energetic metabolism controls key steps of kidney development, homeostasis, and epithelial repair following acute kidney injury (AKI). Hepatocyte nuclear factor-1β (HNF-1β) is a master transcription factor that controls mitochondrial function in proximal tubule (PT) cells. Patients with HNF1B pathogenic variant display a wide range of kidney developmental abnormalities and progressive kidney fibrosis. Characterizing the metabolic changes in PT cells with HNF-1β deficiency may help to identify new targetable molecular hubs involved in HNF1B-related kidney phenotypes and AKI. Here, we combined 1 H-NMR-based metabolomic analysis in a murine PT cell line with CrispR/Cas9-induced Hnf1b invalidation (Hnf1b-/- ), clustering analysis, targeted metabolic assays, and datamining of published RNA-seq and ChIP-seq dataset to identify the role of HNF-1β in metabolism. Hnf1b-/- cells grown in normoxic conditions display intracellular ATP depletion, increased cytosolic lactate concentration, increased lipid droplet content, failure to use pyruvate for energetic purposes, increased levels of tricarboxylic acid (TCA) cycle intermediates and oxidized glutathione, and a reduction of TCA cycle byproducts, all features consistent with mitochondrial dysfunction and an irreversible switch toward glycolysis. Unsupervised clustering analysis showed that Hnf1b-/- cells mimic a hypoxic signature and that they cannot furthermore increase glycolysis-dependent energetic supply during hypoxic challenge. Metabolome analysis also showed alteration of phospholipid biosynthesis in Hnf1b-/- cells leading to the identification of Chka, the gene coding for choline kinase α, as a new putative target of HNF-1β. HNF-1β shapes the energetic metabolism of PT cells and HNF1B deficiency in patients could lead to a hypoxia-like metabolic state precluding further adaptation to ATP depletion following AKI.
Collapse
Affiliation(s)
- Alexis Piedrafita
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Balayssac
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France.,Laboratoire des Interaction Moléculaires et Réactivité Chimique et Photochimique (IMRCP), UMR 5623, Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jean-Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Alexandre Lucas
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Jason S Iacovoni
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Service de Néphrologie, Médecine interne et Hypertension artérielle, Hôpital des Enfants, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Myriam Malet-Martino
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
53
|
Yoshida K, Mushimoto Y, Tanase-Nakao K, Akiba K, Ishii K, Urakami T, Sugihara S, Kikuchi T, Fukami M, Narumi S. A case report with functional characterization of a HNF1B mutation (p.Leu168Pro) causing MODY5. Clin Pediatr Endocrinol 2021; 30:179-185. [PMID: 34629740 PMCID: PMC8481079 DOI: 10.1297/cpe.30.179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/20/2021] [Indexed: 11/25/2022] Open
Abstract
We previously performed next-generation sequencing-based genetic screening in patients
with autoantibody-negative type 1 diabetes, and identified the p.Leu168Pro mutation in
HNF1B. Here,we report the clinical course of the patient and
the results of functional characterization of this mutation. The proband had bilateral
renal hypodysplasia and developed insulin-dependent diabetes during childhood. The
pathogenicity of Leu168Pro-HNF1B was evaluated with three-dimensional structure modeling,
Western blotting, immunofluorescence analysis and luciferase reporter assays using human
embryonic kidney 293 cells. Three-dimensional structure modeling predicted that the Leu168
residue is buried in the DNA-binding Pit-Oct-Unc-specific (POUS) domain and
forms a hydrophobic core. Western blotting showed that the protein expression level of
Leu168Pro-HNF1B was lower than that of wild-type (WT) HNF1B. Immunofluorescence staining
showed that both WT- and Leu168Pro-HNF1B were normally localized in the nucleus. The cells
transfected with WT-HNF1B exhibited 5-fold higher luciferase reporter activity than cells
transfected with an empty vector. The luciferase activities were comparable between
WT-HNF1B/Leu168Pro-HNF1B and WT-HNF1B/empty vector co-transfection. In conclusion,
Leu168Pro is a protein-destabilizing HNF1B mutation, and the
destabilization is likely due to the structural changes involving the hydrophobic core of
POUS. The disease-causing Leu168Pro HNF1B mutation is a
loss-of-function mutation without a dominant-negative effect.
Collapse
Affiliation(s)
- Kei Yoshida
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan.,Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Yuichi Mushimoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kanako Tanase-Nakao
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kazuhisa Akiba
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kanako Ishii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tatsuhiko Urakami
- Department of Pediatrics and Child Health, Nihon University School of Medicine, Tokyo, Japan
| | - Shigetaka Sugihara
- Department of Pediatrics, Tokyo Women's Medical University Medical Center East, Tokyo, Japan
| | - Toru Kikuchi
- Department of Pediatrics, Saitama Medical University, Saitama, Japan
| | - Maki Fukami
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Satoshi Narumi
- Department of Molecular Endocrinology, National Research Institute for Child Health and Development, Tokyo, Japan
| | | |
Collapse
|
54
|
Xiao TL, Zhang J, Liu L, Zhang B. Hepatocyte nuclear factor 1B mutation in a Chinese family with renal cysts and diabetes syndrome: A case report. World J Clin Cases 2021; 9:8461-8469. [PMID: 34754854 PMCID: PMC8554446 DOI: 10.12998/wjcc.v9.i28.8461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 06/18/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Renal cysts and diabetes (RCAD) syndrome is an autosomal dominant diabetic renal disease. Precise molecular diagnosis of RCAD syndrome has proven valuable for understanding its mechanism and personalized therapy.
CASE SUMMARY A RCAD patient and her family were studied to investigate potential responsible genes by the whole exome sequencing (WES). Candidate pathogenic variants were validated by Sanger sequencing. The clinical characteristics of RCAD patient were collected from medical records. Unlike those typical RCAD patients, we observed renal manifestation and prediabetes phenotype, but not reproductive organ phenotype and hypomagnesaemia. A novel 7-bp deletion mutation in exon 4 of the hepatocyte nuclear factor 1B, NM_000458: c.882_888del (p.V294fs), was identified by WES and confirmed by Sanger sequencing.
CONCLUSION This novel mutation identified in a Chinese family with RCAD syndrome might be the molecular pathogenic basis of this disorder.
Collapse
Affiliation(s)
| | - Jun Zhang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Li Liu
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| | - Bo Zhang
- Department of Nephrology, The Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing 400037, China
| |
Collapse
|
55
|
Ruzgienė D, Sutkevičiūtė M, Burnytė B, Grigalionienė K, Jankauskienė A. Reverse Phenotyping Maternal Cystic Kidney Disease by Diagnosis in a Newborn: Case Report and Literature Review on Neonatal Cystic Kidney Diseases. Acta Med Litu 2021; 28:308-316. [PMID: 35474932 PMCID: PMC8958653 DOI: 10.15388/amed.2021.28.2.5] [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: 02/23/2021] [Revised: 04/27/2021] [Accepted: 05/31/2021] [Indexed: 11/22/2022] Open
Abstract
Summary. Kidney cysts are the most common kidney lesion, while congenital kidney cysts are mostly found in pediatric population. Neonatal kidney cysts can develop due to fetal malformations, rare genetic disorders or can be acquired which is very rare. Kidney cysts may be the only isolated finding or be part of the overall phenotype. They can be asymptomatic, found by ultrasound accidentally or can manifest from mild to life-threatening symptoms. Therefore, early diagnosis is very important. Autosomal dominant polycystic kidney disease and autosomal recessive polycystic kidney disease are the most common causes of kidney cysts in the neonatal population. This review highlights the most common kidney cystic diseases during the neonatal period and a rare clinical case of HNF1B-associated disease.
Collapse
|
56
|
Hyperuricemia Is an Early and Relatively Common Feature in Children with HNF1B Nephropathy but Its Utility as a Predictor of the Disease Is Limited. J Clin Med 2021; 10:jcm10153265. [PMID: 34362049 PMCID: PMC8346958 DOI: 10.3390/jcm10153265] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Hyperuricemia is recognized as an important feature of nephropathy, associated with a mutation in the hepatocyte nuclear factor-1B (HNF1B) gene, and could serve as a useful marker of the disease. However, neither a causal relationship nor its predictive value have been proven. The purpose of this study was to assess this in children with renal malformations, both with (mut+) and without HNF1B mutations (mut-). Methods: We performed a retrospective analysis of clinical characteristics of pediatric patients tested for HNF1B mutations, collected in a national registry. Results: 108 children were included in the study, comprising 43 mut+ patients and 65 mut- subjects. Mean sUA was higher and hyperuricemia more prevalent (42.5% vs. 15.4%) in HNF1B carriers. The two groups were similar with respect to respect to age, sex, anthropometric parameters, hypertension, and renal function. Renal function, fractional excretion of uric acid and parathyroid hormone level were independent predictors of sUA. The potential of hyperuricemia to predict mutation was low, and addition of hyperuricemia to a multivariate logistic regression model did not increase its accuracy. Conclusions: Hyperuricemia is an early and common feature of HNF1B nephropathy. A strong association of sUA with renal function and parathyroid hormone limits its utility as a reliable marker to predict HNF1B mutation among patients with kidney anomalies.
Collapse
|
57
|
Maturity Onset Diabetes of the Young-New Approaches for Disease Modelling. Int J Mol Sci 2021; 22:ijms22147553. [PMID: 34299172 PMCID: PMC8303136 DOI: 10.3390/ijms22147553] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 07/04/2021] [Accepted: 07/09/2021] [Indexed: 02/08/2023] Open
Abstract
Maturity-onset diabetes of the young (MODY) is a genetically heterogeneous group of monogenic endocrine disorders that is characterised by autosomal dominant inheritance and pancreatic β-cell dysfunction. These patients are commonly misdiagnosed with type 1 or type 2 diabetes, as the clinical symptoms largely overlap. Even though several biomarkers have been tested none of which could be used as single clinical discriminator. The correct diagnosis for individuals with MODY is of utmost importance, as the applied treatment depends on the gene mutation or is subtype-specific. Moreover, in patients with HNF1A-MODY, additional clinical monitoring can be included due to the high incidence of vascular complications observed in these patients. Finally, stratification of MODY patients will enable better and newer treatment options for MODY patients, once the disease pathology for each patient group is better understood. In the current review the clinical characteristics and the known disease-related abnormalities of the most common MODY subtypes are discussed, together with the up-to-date applied diagnostic criteria and treatment options. Additionally, the usage of pluripotent stem cells together with CRISPR/Cas9 gene editing for disease modelling with the possibility to reveal new pathophysiological mechanisms in MODY is discussed.
Collapse
|
58
|
Patouni K, Cinek O, Pruhova S, Elblova L, Xatzipsalti M, Sertedaki A, Vazeou A. A case of digenic maturity onset diabetes of the young with heterozygous variants in both HNF1Α and HNF1Β genes. Eur J Med Genet 2021; 64:104264. [PMID: 34161864 DOI: 10.1016/j.ejmg.2021.104264] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 05/27/2021] [Accepted: 06/18/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND Maturity onset diabetes of the young (MODY) is the most commonly reported form of monogenic diabetes in the pediatric population. Only a few cases of digenic MODY have been reported up to now. CASE REPORT A female patient was diagnosed with diabetes at the age of 7 years and was treated with insulin. A strong family history of diabetes was present in the maternal side of the family. The patient also presented hypomagnesemia, glomerulocystic kidney disease and a bicornuate uterus. Genetic testing of the patient revealed that she was a double heterozygous carrier of HNF1A gene variant c.685C > T; (p.Arg229Ter) and a whole gene deletion of the HNF1B gene. Her mother was a carrier of the same HNF1A variant. CONCLUSION Digenic inheritance of MODY pathogenic variants is probably more common than currently reported in literature. The use of Next Generation Sequencing panels in testing strategies for MODY could unmask such cases that would otherwise remain undiagnosed.
Collapse
Affiliation(s)
- Konstantina Patouni
- Diabetes Center, First Department of Paediatrics, "P. & A. Kyriakou" Children's Hospital, Athens, Greece.
| | - Ondrej Cinek
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Stepanka Pruhova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Lenka Elblova
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | - Maria Xatzipsalti
- Diabetes Center, First Department of Paediatrics, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| | - Amalia Sertedaki
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Andriani Vazeou
- Diabetes Center, First Department of Paediatrics, "P. & A. Kyriakou" Children's Hospital, Athens, Greece
| |
Collapse
|
59
|
De novo mutation in HNF-1β gene as a cause for Maturity-onset Diabetes of the Young type 5 with sustained hypomagnesemia. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-020-00904-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
60
|
Przepiorski A, Crunk AE, Espiritu EB, Hukriede NA, Davidson AJ. The Utility of Human Kidney Organoids in Modeling Kidney Disease. Semin Nephrol 2021; 40:188-198. [PMID: 32303281 DOI: 10.1016/j.semnephrol.2020.01.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The formation of three-dimensional kidney tissue (organoids) from human pluripotent stem cell lines provides a valuable tool to examine kidney function in an in vitro model and could be used for regenerative medicine approaches. Kidney organoids have the potential to model kidney diseases and congenital defects, be used for drug development, and to further our understanding of acute kidney injury, fibrosis, and chronic kidney disease. In this review, we examine the current stage of pluripotent stem cell-derived kidney organoid technology, challenges, shortcomings, and regenerative potential of kidney organoids in the future.
Collapse
Affiliation(s)
- Aneta Przepiorski
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA.
| | - Amanda E Crunk
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Eugenel B Espiritu
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Neil A Hukriede
- Department of Developmental Biology, University of Pittsburgh, School of Medicine, Pittsburgh, PA; Center for Critical Care Nephrology, University of Pittsburgh, School of Medicine, Pittsburgh, PA
| | - Alan J Davidson
- Department of Molecular Medicine and Pathology, School of Medical Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
61
|
Zhou CX, Zhu XY, Zhu YJ, Gu LL, He LL, Liu W, Yang Y, Wu X, Duan HL, Ru T, Li J. Prenatal features of 17q12 microdeletion and microduplication syndromes: A retrospective case series. Taiwan J Obstet Gynecol 2021; 60:232-237. [PMID: 33678321 DOI: 10.1016/j.tjog.2021.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 10/22/2022] Open
Abstract
OBJECTIVE To present the experience on prenatal features of 17q12 microdeletion and microduplication syndromes. MATERIALS AND METHODS Prenatal chromosomal microarray analysis (CMA) were conducted between January 2015 and December 2018 at a single Chinese tertiary medical centre. Information of cases identified with 17q12 microdeletion or microduplication syndromes were retrospectively collected. Foetal ultrasonographic findings were reviewed, and other information about the gestation week at diagnosis, inheritance and pregnancy outcomes were also included. RESULTS Ten pregnancies with 17q12 microdeletion and 4 with 17q12 microduplication were identified. The copy number variation (CNV) sizes were 1.39-1.94 Mb in the deleted cases and 1.42-1.48 Mb in the duplicated cases, respectively. All the duplicated and deleted regions included HNF1B and LHX1 genes. Most individuals with 17q12 deletion presented kidney anomalies (9/10), with renal hyperechogenicity being the most common finding (7/10). Fetuses with 17q12 duplication presented a wide phenotypic spectrum, including "double bubble" sign, structural anomalies of the heart and growth anomalies. CONCLUSIONS Our experience further demonstrated the high correlation between 17q12 microdeletion and renal anomalies especially hyperechogenic kidneys. Structural anomalies of the heart were newly identified phenotypes of 17q12 duplication during prenatal period. Besides, growth anomalies and duodenal atresia might be associated with the duplication.
Collapse
Affiliation(s)
- Chun-Xiang Zhou
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Xiang-Yu Zhu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Yu-Jie Zhu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Lei-Lei Gu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Lin-Lin He
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Wei Liu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Ying Yang
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Xing Wu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Hong-Lei Duan
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Tong Ru
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Jie Li
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
62
|
Motyka R, Kołbuc M, Wierzchołowski W, Beck BB, Towpik IE, Zaniew M. Four Cases of Maturity Onset Diabetes of the Young (MODY) Type 5 Associated with Mutations in the Hepatocyte Nuclear Factor 1 Beta (HNF1B) Gene Presenting in a 13-Year-Old Boy and in Adult Men Aged 33, 34, and 35 Years in Poland. AMERICAN JOURNAL OF CASE REPORTS 2021; 22:e928994. [PMID: 33526762 PMCID: PMC7869582 DOI: 10.12659/ajcr.928994] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Case series Patients: Male, 13-year-old • Male, 33-year-old • Male, 34-year-old • Male, 35-year-old Final Diagnosis: HNF1B nephropathy • HNF1B-MODY type 5 Symptoms: Diabetic ketoacidosis • elevated liver enzymes • hyperglycemia • hyperuricemia • hypomagnesemia • positive family history • renal cysts • renal magnesium wasting • weigh loss Medication:— Clinical Procedure: Genetic analysis • islet autoantibodies Specialty: Endocrinology and Metabolic • Nephrology
Collapse
Affiliation(s)
- Rafał Motyka
- University of Zielona Góra, Zielona Góra, Poland
| | - Marcin Kołbuc
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | | | - Bodo B Beck
- Institute of Human Genetics and Center for Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Iwona Ewa Towpik
- Department of Internal Medicine, Univiersity of Zielona Góra, Zielona Góra, Poland
| | - Marcin Zaniew
- Department of Pediatrics, Univiersity of Zielona Góra, Zielona Góra, Poland
| |
Collapse
|
63
|
Analysis of expression, epigenetic, and genetic changes of HNF1B in 130 kidney tumours. Sci Rep 2020; 10:17151. [PMID: 33051485 PMCID: PMC7555858 DOI: 10.1038/s41598-020-74059-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/23/2020] [Indexed: 12/18/2022] Open
Abstract
Hepatocyte nuclear factor 1 beta (HNF1B) is a transcription factor which plays a crucial role in nephronogenesis, and its germline mutations have been associated with kidney developmental disorders. However, the effects of HNF1B somatic exonic mutations and its role in the pathogenesis of kidney tumours has not yet been elucidated. Depending on the type of the tumour HNF1B may act as a tumour suppressor or oncogene, although the exact mechanism by which HNF1B participates in the process of cancerogenesis is unknown. Using an immunohistochemical approach, and methylation and mutation analysis, we have investigated the expression, epigenetic, and genetic changes of HNF1B in 130 cases of renal tumours (121 renal cell carcinomas, 9 oncocytomas). In the subset of clear cell renal cell carcinoma (ccRCC), decreased HNF1B expression was associated with a higher tumour grade and higher T stage. The mutation analysis revealed no mutations in the analysed samples. Promoter methylation was detected in two ccRCCs and one oncocytoma. The results of our work on a limited sample set suggest that while in papillary renal cell carcinoma HNF1B functions as an oncogene, in ccRCC and chRCC it may act in a tumour suppressive fashion.
Collapse
|
64
|
Izzi C, Dordoni C, Econimo L, Delbarba E, Grati FR, Martin E, Mazza C, Savoldi G, Rampoldi L, Alberici F, Scolari F. Variable Expressivity of HNF1B Nephropathy, From Renal Cysts and Diabetes to Medullary Sponge Kidney Through Tubulo-interstitial Kidney Disease. Kidney Int Rep 2020; 5:2341-2350. [PMID: 33305128 PMCID: PMC7710890 DOI: 10.1016/j.ekir.2020.09.042] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/24/2020] [Accepted: 09/22/2020] [Indexed: 12/18/2022] Open
Abstract
Introduction In humans, heterozygous mutations of hepatocyte nuclear factor 1beta (HNF1B) are responsible for a dominant inherited disease with both renal and extrarenal phenotypes. HNF1B nephropathy is the umbrella term that includes the various kidney phenotypes of the disease, ranging from congenital anomalies of the kidney and urinary tract (CAKUT), to tubular transport abnormalities, to chronic tubulointerstitial and cystic renal disease. Methods We describe 7 families containing 13 patients with ascertained HNF1B nephropathy. All patients underwent genetic testing and clinical, laboratory, and instrumental assessment, including renal imaging and evaluation of extrarenal HNF1B manifestations. Results Significant inter- and intrafamilial variability of HNF1B nephropathy has been observed. In our cohort, HNF1B pathogenic variants presented with renal cysts and diabetes syndrome (RCAD); renal cystic phenotype mimicking autosomal dominant polycystic kidney disease (ADPKD); autosomal dominant tubulointerstitial kidney disease (ADTKD) with or without hyperuricemia and gout; CAKUT; and nephrogenic diabetes insipidus (NDI). Of note, for the first time, we describe the occurrence of medullary sponge kidney (MSK) in a family harboring the HNF1B whole-gene deletion at chromosome 17q12. Genotype characterization led to the identification of an additional 6 novel HNF1B pathogenic variants, 3 frameshift, 2 missense, and 1 nonsense. Conclusion HNF1B nephropathy may present with a highly variable renal phenotype in adult patients. We expand the HNF1B renal clinical picture to include MSK as a potential new finding. Finally, we expand the allelic repertoire of the disease by adding novel HNF1B pathogenic variants.
Collapse
Affiliation(s)
- Claudia Izzi
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia Italy.,Prenatal Diagnosis Unit, Department of Obstetrics and Gynecology, ASST Spedali Civili, Brescia, Italy
| | - Chiara Dordoni
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia Italy.,Prenatal Diagnosis Unit, Department of Obstetrics and Gynecology, ASST Spedali Civili, Brescia, Italy
| | - Laura Econimo
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia Italy
| | - Elisa Delbarba
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia Italy
| | - Francesca Romana Grati
- Cytogenetics and Medical Genetics Unit TOMA Advanced Biomedical Assays (Impact Lab Group), Busto Arsizio, Italy
| | - Eva Martin
- Radiology Unit, Montichiari Hospital, ASST Spedali Civili, Brescia, Italy
| | - Cinzia Mazza
- Medical Genetics Laboratory, ASST-Spedali Civili, Brescia, Italy
| | | | - Luca Rampoldi
- Molecular Genetics of Renal Disorders, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federico Alberici
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia Italy
| | - Francesco Scolari
- Division of Nephrology and Dialysis, Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia and ASST Spedali Civili of Brescia, Brescia Italy
| |
Collapse
|
65
|
Bonnefond A, Boissel M, Bolze A, Durand E, Toussaint B, Vaillant E, Gaget S, Graeve FD, Dechaume A, Allegaert F, Guilcher DL, Yengo L, Dhennin V, Borys JM, Lu JT, Cirulli ET, Elhanan G, Roussel R, Balkau B, Marre M, Franc S, Charpentier G, Vaxillaire M, Canouil M, Washington NL, Grzymski JJ, Froguel P. Pathogenic variants in actionable MODY genes are associated with type 2 diabetes. Nat Metab 2020; 2:1126-1134. [PMID: 33046911 DOI: 10.1038/s42255-020-00294-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
Genome-wide association studies have identified 240 independent loci associated with type 2 diabetes (T2D) risk, but this knowledge has not advanced precision medicine. In contrast, the genetic diagnosis of monogenic forms of diabetes (including maturity-onset diabetes of the young (MODY)) are textbook cases of genomic medicine. Recent studies trying to bridge the gap between monogenic diabetes and T2D have been inconclusive. Here, we show a significant burden of pathogenic variants in genes linked with monogenic diabetes among people with common T2D, particularly in actionable MODY genes, thus implying that there should be a substantial change in care for carriers with T2D. We show that, among 74,629 individuals, this burden is probably driven by the pathogenic variants found in GCK, and to a lesser extent in HNF4A, KCNJ11, HNF1B and ABCC8. The carriers with T2D are leaner, which evidences a functional metabolic effect of these mutations. Pathogenic variants in actionable MODY genes are more frequent than was previously expected in common T2D. These results open avenues for future interventions assessing the clinical interest of these pathogenic mutations in precision medicine.
Collapse
Affiliation(s)
- Amélie Bonnefond
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| | - Mathilde Boissel
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | | | - Emmanuelle Durand
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Bénédicte Toussaint
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Emmanuel Vaillant
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Stefan Gaget
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Franck De Graeve
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Aurélie Dechaume
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Frédéric Allegaert
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - David Le Guilcher
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Loïc Yengo
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
- Institute for Molecular Bioscience, the University of Queensland, St Lucia, Australia
| | - Véronique Dhennin
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | | | | | | | - Gai Elhanan
- Desert Research Institute, Reno, NV, USA
- Renown Institute of Health Innovation, Reno, NV, USA
| | - Ronan Roussel
- Department of Diabetology Endocrinology Nutrition, Hôpital Bichat, DHU FIRE, Assistance Publique Hôpitaux de Paris, Paris, France
- Inserm U1138, Centre de Recherche des Cordeliers, Paris, France
- UFR de Médecine, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Beverley Balkau
- Inserm U1018, Institut Gustave Roussy, Center for Research in Epidemiology and Population Health, Villejuif, France
- University Paris-Saclay, University Paris-Sud, Villejuif, France
| | - Michel Marre
- Inserm U1138, Centre de Recherche des Cordeliers, Paris, France
- CMC Ambroise Paré, Neuilly-sur-Seine, France
| | - Sylvia Franc
- CERITD (Centre d'Étude et de Recherche pour l'Intensification du Traitement du Diabète), Evry, France
- Department of Diabetes, Sud-Francilien Hospital, University Paris-Sud, Orsay, Corbeil-Essonnes, France
| | - Guillaume Charpentier
- CERITD (Centre d'Étude et de Recherche pour l'Intensification du Traitement du Diabète), Evry, France
| | - Martine Vaxillaire
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | - Mickaël Canouil
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France
| | | | - Joseph J Grzymski
- Desert Research Institute, Reno, NV, USA
- Renown Institute of Health Innovation, Reno, NV, USA
| | - Philippe Froguel
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Université de Lille, Institut Pasteur de Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Imperial College London, London, UK.
| |
Collapse
|
66
|
Chang K, Wei Z, Cao H. miR-375-3p inhibits the progression of laryngeal squamous cell carcinoma by targeting hepatocyte nuclear factor-1β. Oncol Lett 2020; 20:80. [PMID: 32863913 PMCID: PMC7436894 DOI: 10.3892/ol.2020.11941] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 05/21/2020] [Indexed: 12/16/2022] Open
Abstract
Laryngeal squamous cell carcinoma (LSCC) is one of the most frequently diagnosed head and neck cancers worldwide. Increasing evidence suggests that microRNAs (miRNAs/miRs) regulate the progression of tumorigenesis and the malignant behaviors of cancer cells. The aim of this study was to investigate the function and underlying mechanism of miR-375-3p in LSCC. The expression of miR-375-3p in LSCC tissues and cells was detected using reverse transcription-quantitative PCR. The effects of miR-375-3p on the malignant phenotype of LSCC cells was determined using the Cell Counting Kit-8 assay and flow cytometry. The targets of miR-375-3p were predicted using the miRDB database and confirmed by the luciferase reporter assay. The results of the present study demonstrated that miR-375-3p was downregulated in LSCC tissues and cell lines. Furthermore, overexpression of miR-375-3p significantly suppressed the proliferation and cell cycle progression of LSCC cells. Overexpression of miR-375-3p also increased LSCC cell apoptosis. Mechanistical analysis indicated that miR-375-3p bound the 3′-untranslated region of the hepatocyte nuclear factor 1β (HNF1β) and decreased its expression in LSCC cells. Consistent with the role of HNF1β in glucose metabolism, overexpression of miR-375-3p significantly inhibited glucose consumption and lactate production in LSCC cells. Transfection with HNF1β notably reversed the inhibitory effect of miR-375-3p on the proliferation of LSCC cells. Collectively, these results indicate the tumor suppressive role of miR-375-3p in LSCC via HNF1β, suggesting that miR-375-3p may serve as a potential target in the treatment of LSCC.
Collapse
Affiliation(s)
- Kunpeng Chang
- Department of Otolaryngology Head and Neck Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Zhenxing Wei
- Department of Otolaryngology Head and Neck Surgery, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471000, P.R. China
| | - Hua Cao
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
67
|
Mateus JC, Rivera C, O’Meara M, Valenzuela A, Lizcano F. Maturity-onset diabetes of the young type 5 a MULTISYSTEMIC disease: a CASE report of a novel mutation in the HNF1B gene and literature review. Clin Diabetes Endocrinol 2020; 6:16. [PMID: 32864159 PMCID: PMC7448977 DOI: 10.1186/s40842-020-00103-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetes mellitus with autosomal dominant inheritance, such as maturity-onset diabetes of the young (MODY), is a genetic form of diabetes mellitus. MODY is a type of monogenic diabetes mellitus in which multiple genetic variants may cause an alteration to the functioning of beta cells. The three most known forms of MODY are caused by modifications to the hnf4a, gck, and hnf1a genes. However, other MODY variants can cause multiple alterations in the embryonic development of the endoderm. This is the case in patients presenting with MODY5, who have a mutation of the hepatic nuclear factor 1B (hnf1b) gene. CASE PRESENTATION We present the clinical case of a 15 year-old patient with a family history of diabetes mellitus and a classical MODY type 5 (MODY5) phenotype involving the pancreas and kidney, with a novel, unreported mutation in the hnf1b gene. CONCLUSIONS MODY5 is characterised by a mutation in the hnf1b gene, which plays an important role in the development and function of multiple organs. It should be suspected in patients with unusual diabetes and multisystem involvement unrelated to diabetes. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Juan Camilo Mateus
- Endocrinology Fellowship, School of Medicine and Health Sciences, Rosary University – Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Carolina Rivera
- Department of Genetics, Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Miguel O’Meara
- Department of Endocrinology, Diabetes and Nutrition, Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Alex Valenzuela
- Department of Endocrinology, Diabetes and Nutrition, Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Fernando Lizcano
- Department of Endocrinology, Diabetes and Nutrition, Fundacion Cardio-Infantil IC, Bogotá, Colombia
- Center of Biomedical Investigation Universidad de La Sabana, CIBUS, Chia, CU 250008 Colombia
| |
Collapse
|
68
|
Tao T, Yang Y, Hu Z. A novel HNF1B mutation p.R177Q in autosomal dominant tubulointerstitial kidney disease and maturity-onset diabetes of the young type 5: A pedigree-based case report. Medicine (Baltimore) 2020; 99:e21438. [PMID: 32756155 PMCID: PMC7402722 DOI: 10.1097/md.0000000000021438] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
RATIONALE Mutations in the hepatocyte nuclear factor-1-beta (HNF1B) gene result in a very variable presentation, including maturity onset diabetes of the young (MODY), renal cysts, renal dysplasia, and autosomal dominant tubulointerstitial kidney disease (ADTKD), which is characterized by tubular damage, renal fibrosis, and progressive renal dysfunction. PATIENT CONCERNS A 22-year-old man came to the hospital presenting with hyperglycemia, hyperuricemia and elevated serum creatinine. His urine protein was within the normal range. The ultrasound examination revealed shrunken kidneys with renal cysts. The patient's mother was diagnosed with diabetes mellitus when she was 25 years old. Her laboratory results showed elevated serum creatinine. Her ultrasonography revealed shrunken kidneys with renal cysts and hydronephrosis without kidney stones. The next-generation sequencing revealed that the proband and his mother held the same heterozygous missense mutation (c.530G>A, NM_000458, p.R177Q) in the HNF1B gene. Bioinformatic analyses predicted that the mutation was likely pathogenic. DIAGNOSIS The patient and his mother were diagnosed as ADTKD and MODY5 due to HNF1B mutation. INTERVENTION The proband was administered metformin at a dose of 500 mg/day. OUTCOMES The patient had well-controlled blood glucose levels and a stable renal function at his 12-month follow-up. LESSONS We should take into account the diagnoses of ADTKD and MODY5 if patients present with early onset diabetes and multiple renal cysts or evidence of renal tubulointerstitial dysplasia, especially those with negative proteinuria results. Genetic testing helps detect the HNF1B gene mutations.
Collapse
MESH Headings
- Aftercare
- Central Nervous System Diseases/complications
- Central Nervous System Diseases/drug therapy
- Central Nervous System Diseases/genetics
- Dental Enamel/abnormalities
- Diabetes Mellitus, Type 2/complications
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/genetics
- Hepatocyte Nuclear Factor 1-beta/genetics
- Heterozygote
- High-Throughput Nucleotide Sequencing/methods
- Humans
- Hyperglycemia/etiology
- Hyperuricemia/etiology
- Hypoglycemic Agents/administration & dosage
- Hypoglycemic Agents/therapeutic use
- Kidney/diagnostic imaging
- Kidney/pathology
- Kidney/physiopathology
- Kidney Diseases, Cystic/complications
- Kidney Diseases, Cystic/drug therapy
- Kidney Diseases, Cystic/genetics
- Kidney Diseases, Cystic/pathology
- Male
- Metformin/administration & dosage
- Metformin/therapeutic use
- Mutation, Missense
- Nephritis, Interstitial/complications
- Nephritis, Interstitial/genetics
- Nephritis, Interstitial/pathology
- Pedigree
- Ultrasonography/methods
- Young Adult
Collapse
Affiliation(s)
| | - Yuan Yang
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | |
Collapse
|
69
|
Genotype and Phenotype Analyses in Pediatric Patients with HNF1B Mutations. J Clin Med 2020; 9:jcm9072320. [PMID: 32708349 PMCID: PMC7408390 DOI: 10.3390/jcm9072320] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/15/2020] [Accepted: 07/17/2020] [Indexed: 01/01/2023] Open
Abstract
HNF1B mutations, one of the most common causes of congenital anomalies of the kidney and urinary tract, manifest as various renal and extrarenal phenotypes. We analyzed the genotype-phenotype correlations in 14 pediatric patients with HNF1B mutations. Genetic studies revealed total gene deletion in six patients (43%). All patients had bilateral renal abnormalities, primarily multiple renal cysts. Twelve patients exhibited progressive renal functional deterioration, and six of them progressed to kidney failure. The annual reduction in estimated glomerular filtration rate was−2.1 mL/min/1.73 m2. Diabetes developed in five patients (36%), including one patient with new-onset diabetes after transplantation. Neurological deficits were noted in three patients (21%), one with total gene deletion and two with missense mutations. Pancreatic abnormalities were more frequent in patients with missense mutations than in patients with other types of mutations. Genotype showed no significant correlation with renal outcomes or other extrarenal manifestations. The HNF1B scores at the times of onset and genetic diagnosis were <8 in two patients and one patient, respectively. Diagnosis of HNF1B mutations is clinically difficult because of extreme phenotypic variability and incomplete penetrance. Furthermore, some phenotypes develop with age. Therefore, patient age should be taken into consideration to increase the diagnostic rate, because some phenotypes develop with age.
Collapse
|
70
|
Syndromes de Bartter–Gitelman. Nephrol Ther 2020; 16:233-243. [DOI: 10.1016/j.nephro.2020.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
71
|
Results of targeted next-generation sequencing in children with cystic kidney diseases often change the clinical diagnosis. PLoS One 2020; 15:e0235071. [PMID: 32574212 PMCID: PMC7310724 DOI: 10.1371/journal.pone.0235071] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 06/07/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic kidney diseases are a very heterogeneous group of chronic kidney diseases. The diagnosis is usually based on clinical and ultrasound characteristics and the final diagnosis is often difficult to be made. Next-generation sequencing (NGS) may help the clinicians to find the correct final diagnosis. The aim of our study was to test the diagnostic yield of NGS and its ability to improve the diagnosis precision in a heterogeneous group of children with cystic kidney diseases. Next-generation sequencing of genes responsible for the formation of cystic kidneys was performed in 31 unrelated patients with various clinically diagnosed cystic kidney diseases gathered at the Department of Pediatrics of Motol University Hospital in Prague between 2013 and 2018. The underlying pathogenic variants were detected in 71% of patients (n = 22), no or only one (in case of autosomal recessive inheritance) pathogenic variant was found in 29% of patients (n = 9). The result of NGS correlated with the clinical diagnosis made before the NGS in 55% of patients (n = 17), in the remaining 14 children (45%) the result of NGS revealed another type of cystic kidney disease that was suspected clinically before or did not find causal mutation in suspected genes. The most common unexpected findings were variants in nephronophthisis (NPHP) genes in children with clinically suspected autosomal recessive polycystic kidney disease (ARPKD, n = 4). Overall, 24 pathogenic or probably pathogenic variants were detected in the PKHD1 gene, 8 variants in the TMEM67 gene, 4 variants in the PKD1 gene, 2 variants in the HNF1B gene and 2 variants in BBS1 and NPHP1 genes, respectively. NGS is a valuable tool in the diagnostics of various forms of cystic kidney diseases. Its results changed the clinically based diagnoses in 16% (n = 5) of the children.
Collapse
|
72
|
Berberich AJ, Wang J, Cao H, McIntyre AD, Spaic T, Miller DB, Stock S, Huot C, Stein R, Knoll J, Yang P, Robinson JF, Hegele RA. Simplifying Detection of Copy-Number Variations in Maturity-Onset Diabetes of the Young. Can J Diabetes 2020; 45:71-77. [PMID: 33011132 DOI: 10.1016/j.jcjd.2020.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/29/2020] [Accepted: 06/01/2020] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Copy-number variations (CNVs) are large-scale deletions or duplications of DNA that have required specialized detection methods, such as microarray-based genomic hybridization or multiplex ligation probe amplification. However, recent advances in bioinformatics have made it possible to detect CNVs from next-generation DNA sequencing (NGS) data. Maturity-onset diabetes of the young (MODY) 5 is a subtype of autosomal-dominant diabetes that is often caused by heterozygous deletions involving the HNF1B gene on chromosome 17q12. We evaluated the utility of bioinformatic processing of raw NGS data to detect chromosome 17q12 deletions in MODY5 patients. METHODS NGS data from 57 patients clinically suspected to have MODY but who were negative for pathogenic mutations using a targeted panel were re-examined using a CNV calling tool (CNV Caller, VarSeq version 1.4.3). Potential CNVs for MODY5 were then confirmed using whole-exome sequencing, cytogenetic analysis and breakpoint analysis when possible. RESULTS Whole-gene deletions in HNF1B, ranging from 1.46 to 1.85 million basepairs in size, were detected in 3 individuals with features of MODY5. These were confirmed by independent methods to be part of a more extensive 17q12 deletion syndrome. Two additional patients carrying a 17q12 deletion were subsequently diagnosed using this method. CONCLUSIONS Large-scale deletions are the most common cause of MODY5 and can be detected directly from NGS data, without the need for additional methods.
Collapse
Affiliation(s)
- Amanda J Berberich
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| | - Jian Wang
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Henian Cao
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Adam D McIntyre
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Tamara Spaic
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - David B Miller
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Suzanne Stock
- Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Celine Huot
- Department of Pediatrics, CHU Sainte-Justine, University of Montreal, Montréal, Quebec, Canada
| | - Robert Stein
- Department of Pediatrics, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Joan Knoll
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Ping Yang
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - John F Robinson
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A Hegele
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
73
|
Wang J, Wang Y, Wang L, Chen WY, Sheng M. The diagnostic yield of intellectual disability: combined whole genome low-coverage sequencing and medical exome sequencing. BMC Med Genomics 2020; 13:70. [PMID: 32429945 PMCID: PMC7236547 DOI: 10.1186/s12920-020-0726-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/29/2020] [Indexed: 02/06/2023] Open
Abstract
Background Intellectual disability (ID) is a heterogeneous neurodevelopmental disorder with a complex genetic underpinning in its etiology. Chromosome microarray (CMA) is recommended as the first-tier diagnostic test for ID due to high detection rate of copy number variation (CNV). Methods To identify an appropriate clinical detection scheme for ID in Han Chinese patients, whole genome low-coverage sequencing was performed as the first-tier diagnostic test, and medical exome sequencing (MES) as the second-tier diagnostic test for patients with negative results of CNVs. Results A total of 19 pathogenic CNVs in 16/95(16.84%) ID patients and 10 pathogenic single-nucleotide variations (SNVs), including 6 novel mutations in 8/95(8.42%) ID patients were identified on whom no pathogenic CNVs were discovered. The detection rate of CNVs in ID with multiple congenital anomalies (MCA) subgroup was significantly higher than ID with autism spectrum disorders and other IDs subgroups. And the single-nucleotide variations showed a higher occurrence rate in the other IDs subgroup. Conclusions There were differences in the diagnostic yields of different variation types among the three ID subgroups. Our findings provided a new perspective on appropriate clinical detection scheme in different ID subgroups based on statistically significant differences among the three ID subgroups. The application of whole genome low-coverage sequencing as the first-tier diagnostic test for ID with MCA subgroup and MES as the first-tier diagnostic test for other ID subgroup was considered as an efficient clinical detection scheme.
Collapse
Affiliation(s)
- Jun Wang
- Department of Neurology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Yan Wang
- Department of Neurology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, 100020, China
| | - Liwen Wang
- Department of Neurology, Affiliated Children's Hospital of Capital Institute of Pediatrics, Beijing, 100020, China
| | - Wang Yang Chen
- Kaiumph Medical Diagnostics Co,Ltd, Beijing, 100102, China
| | - Min Sheng
- Kaiumph Medical Diagnostics Co,Ltd, Beijing, 100102, China
| |
Collapse
|
74
|
Xu A, Lin Y, Sheng H, Cheng J, Mei H, Ting TH, Zeng C, Liang C, Zhang W, Li C, Li X, Liu L. Molecular diagnosis of maturity-onset diabetes of the young in a cohort of Chinese children. Pediatr Diabetes 2020; 21:431-440. [PMID: 31957151 DOI: 10.1111/pedi.12985] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 12/12/2019] [Accepted: 01/13/2020] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The purpose of this study was to investigate the molecular basis of maturity-onset diabetes of the young (MODY) by whole-exome sequencing (WES) and estimate the frequency and describe the clinical characteristics of MODY in southern China. METHODS Genetic analysis was performed in 42 patients with MODY aged 1 month to 18 years among a cohort of 759 patients with diabetes, identified with the following four clinical criteria: age of diagnosis ≤18 years; negative pancreatic autoantibodies; family history of diabetes; or persistently detectable C-peptide; or diabetes associated with extrapancreatic features. GCK gene mutations were first screened by Sanger sequencing. GCK mutation-negative patients were further analyzed by WES. RESULTS Mutations were identified in 24 patients: 20 mutations in GCK, 1 in HNF4A, 1 in INS, 1 in ABCC8, and a 17q12 microdeletion. Four previously unpublished novel GCK mutations: c.1108G>C in exon 9, and c.1339C>T, c.1288_1290delCTG, and c.1340_1343delGGGGinsCTGGTCT in exon 10 were detected. WES identified a novel missense mutation c.311A>G in exon 3 in the INS gene, and copy number variation analysis detected a 1.4 Mb microdeletion in the long arm of the chromosome 17q12 region. Compared with mutation-negative subjects, the mutation-positive subjects had lower hemoglobin A1c and initial blood glucose levels. CONCLUSIONS Most MODY cases in this study were due to GCK mutations, which is in contrast to previous reports in Chinese patients. Diabetes associated with extrapancreatic features should be a clinical criterion for MODY genetic analysis. Mutational analysis by WES provided a precise diagnosis of MODY subtypes. Moreover, WES can be useful for detecting large deletions in coding regions in addition to point mutations.
Collapse
Affiliation(s)
- Aijing Xu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yunting Lin
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huiying Sheng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Jing Cheng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Huifen Mei
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Tzer Hwu Ting
- Department of Paediatrics, Faculty of Medicine & Health Sciences, University Putra Malaysia, Serdang, Malaysia
| | - Chunhua Zeng
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Cuili Liang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Wen Zhang
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Cuiling Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiuzhen Li
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Liu
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
75
|
Riedhammer KM, Braunisch MC, Günthner R, Wagner M, Hemmer C, Strom TM, Schmaderer C, Renders L, Tasic V, Gucev Z, Nushi-Stavileci V, Putnik J, Stajić N, Weidenbusch M, Uetz B, Montoya C, Strotmann P, Ponsel S, Lange-Sperandio B, Hoefele J. Exome Sequencing and Identification of Phenocopies in Patients With Clinically Presumed Hereditary Nephropathies. Am J Kidney Dis 2020; 76:460-470. [PMID: 32359821 DOI: 10.1053/j.ajkd.2019.12.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 12/15/2019] [Indexed: 12/11/2022]
Abstract
RATIONALE & OBJECTIVE Hereditary nephropathies are clinically and genetically heterogeneous disorders. For some patients, the clinical phenotype corresponds to a specific hereditary disease but genetic testing reveals that the expected genotype is not present (phenocopy). The aim of this study was to evaluate the spectrum and frequency of phenocopies identified by using exome sequencing in a cohort of patients who were clinically suspected to have hereditary kidney disorders. STUDY DESIGN Cross-sectional cohort study. SETTING & PARTICIPANTS 174 unrelated patients were recruited for exome sequencing and categorized into 7 disease groups according to their clinical presentation. They included autosomal dominant tubulointerstitial kidney disease, Alport syndrome, congenital anomalies of the kidney and urinary tract, ciliopathy, focal segmental glomerulosclerosis/steroid-resistant nephrotic syndrome, VACTERL association, and "other." RESULTS A genetic diagnosis (either likely pathogenic or pathogenic variant according to the guidelines of the American College of Medical Genetics) was established using exome sequencing in 52 of 174 (30%) cases. A phenocopy was identified for 10 of the 52 exome sequencing-solved cases (19%), representing 6% of the total cohort. The most frequent phenocopies (n=5) were associated with genetic Alport syndrome presenting clinically as focal segmental glomerulosclerosis/steroid-resistant nephrotic syndrome. Strictly targeted gene panels (<25 kilobases) did not identify any of the phenocopy cases. LIMITATIONS The spectrum of described phenocopies is small. Selection bias may have altered the diagnostic yield within disease groups in our study population. The study cohort was predominantly of non-Finnish European descent, limiting generalizability. Certain hereditary kidney diseases cannot be diagnosed by using exome sequencing (eg, MUC1-autosomal dominant tubulointerstitial kidney disease). CONCLUSIONS Phenocopies led to the recategorization of disease and altered clinical management. This study highlights that exome sequencing can detect otherwise occult genetic heterogeneity of kidney diseases.
Collapse
Affiliation(s)
- Korbinian M Riedhammer
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany; Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matthias C Braunisch
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany; Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Roman Günthner
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany; Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Matias Wagner
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany; Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Clara Hemmer
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Tim M Strom
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Christoph Schmaderer
- Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Lutz Renders
- Department of Nephrology, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Velibor Tasic
- University Children's Hospital, Medical Faculty of Skopje, Macedonia
| | - Zoran Gucev
- University Children's Hospital, Medical Faculty of Skopje, Macedonia
| | | | - Jovana Putnik
- Institute for Mother and Child Health Care of Serbia "Dr Vukan Čupić", Department of Nephrology, University of Belgrade, Faculty of Medicine, Belgrade, Serbia
| | - Nataša Stajić
- Institute for Mother and Child Health Care of Serbia "Dr Vukan Čupić", Department of Nephrology, University of Belgrade, Faculty of Medicine, Belgrade, Serbia
| | - Marc Weidenbusch
- Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Ludwig-Maximilians University, Munich, Germany
| | - Barbara Uetz
- München-Klinik Schwabing, Klinikum rechts der Isar, Technical University of Munich, Children's Hospital, Pediatric Nephrology, Munich, Germany; KfH-Kindernierenzentrum, Munich, Germany
| | | | - Peter Strotmann
- München-Klinik Schwabing, Klinikum rechts der Isar, Technical University of Munich, Children's Hospital, Pediatric Nephrology, Munich, Germany
| | - Sabine Ponsel
- Division of Pediatric Nephrology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Baerbel Lange-Sperandio
- Division of Pediatric Nephrology, Dr. v. Hauner Children's Hospital, Ludwig-Maximilians University, Munich, Germany
| | - Julia Hoefele
- Institute of Human Genetics, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
76
|
Adalat S, Hayes WN, Bryant WA, Booth J, Woolf AS, Kleta R, Subtil S, Clissold R, Colclough K, Ellard S, Bockenhauer D. HNF1B Mutations Are Associated With a Gitelman-like Tubulopathy That Develops During Childhood. Kidney Int Rep 2019; 4:1304-1311. [PMID: 31517149 PMCID: PMC6732753 DOI: 10.1016/j.ekir.2019.05.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/29/2019] [Accepted: 05/20/2019] [Indexed: 12/20/2022] Open
Abstract
Background Mutations in the transcription factor hepatocyte nuclear factor 1B (HNF1B) are the most common inherited cause of renal malformations, yet also associated with renal tubular dysfunction, most prominently magnesium wasting with hypomagnesemia. The presence of hypomagnesemia has been proposed to help select appropriate patients for genetic testing. Yet, in a large cohort, hypomagnesemia was discriminatory only in adult, but not in pediatric patients. We therefore investigated whether hypomagnesemia and other biochemical changes develop with age. Methods We performed a retrospective analysis of clinical, biochemical, and genetic results of pediatric patients with renal malformations tested for HNF1B mutations, separated into 4 age groups. Values were excluded if concurrent estimated glomerular filtration rate (eGFR) was <30 ml/min per 1.73 m2, or after transplantation. Results A total of 199 patients underwent HNF1B genetic testing and mutations were identified in 52 (mut+). The eGFRs were comparable between mut+ and mut- in any age group. Although median plasma magnesium concentrations differed significantly between mut+ and mut- patients in all age groups, overt hypomagnesemia was not present until the second half of childhood in the mut+ group. There was also a significant difference in median potassium concentrations in late childhood with lower values in the mut+ cohort. Conclusions The abnormal tubular electrolyte handling associated with HNF1B mutations develops with age and is not restricted to magnesium, but consistent with a more generalized dysfunction of the distal convoluted tubule, reminiscent of Gitelman syndrome. The absence of these abnormalities in early childhood should not preclude HNF1B mutations from diagnostic considerations.
Collapse
Affiliation(s)
- Shazia Adalat
- Evelina Children’s Hospital, London, United Kingdom
- UCL Department of Renal Medicine, London, United Kingdom
| | - Wesley N. Hayes
- UCL Department of Renal Medicine, London, United Kingdom
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - William A. Bryant
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - John Booth
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | - Adrian S. Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, United Kingdom
- Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - Robert Kleta
- UCL Department of Renal Medicine, London, United Kingdom
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
| | | | - Rhian Clissold
- Royal Devon and Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Kevin Colclough
- Department of Molecular Genetics, Royal Devon & Exeter NHS Foundation Trust, Exeter, United Kingdom
| | - Sian Ellard
- Department of Molecular Genetics, Royal Devon & Exeter NHS Foundation Trust, Exeter, United Kingdom
- Institute of Biomedical and Clinical Science, College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Detlef Bockenhauer
- UCL Department of Renal Medicine, London, United Kingdom
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, United Kingdom
- Correspondence: Detlef Bockenhauer, UCL Department of Renal Medicine, London WC1N 3JH, United Kingdom.
| |
Collapse
|
77
|
Nagano C, Morisada N, Nozu K, Kamei K, Tanaka R, Kanda S, Shiona S, Araki Y, Ohara S, Matsumura C, Kasahara K, Mori Y, Seo A, Miura K, Washiyama M, Sugimoto K, Harada R, Tazoe S, Kourakata H, Enseki M, Aotani D, Yamada T, Sakakibara N, Yamamura T, Minamikawa S, Ishikura K, Ito S, Hattori M, Iijima K. Clinical characteristics of HNF1B-related disorders in a Japanese population. Clin Exp Nephrol 2019; 23:1119-1129. [PMID: 31131422 DOI: 10.1007/s10157-019-01747-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/07/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hepatocyte nuclear factor 1β (HNF1B), located on chromosome 17q12, causes renal cysts and diabetes syndrome (RCAD). Moreover, various phenotypes related to congenital anomalies of the kidney and urinary tract (CAKUT) or Bartter-like electrolyte abnormalities can be caused by HNF1B variants. In addition, 17q12 deletion syndrome presents with multi-system disorders, as well as RCAD. As HNF1B mutations are associated with different phenotypes and genotype-phenotype relationships remain unclear, here, we extensively studied these mutations in Japan. METHODS We performed genetic screening of RCAD, CAKUT, and Bartter-like syndrome cases. Heterozygous variants or whole-gene deletions in HNF1B were detected in 33 cases (19 and 14, respectively). All deletion cases were diagnosed as 17q12 deletion syndrome, confirmed by multiplex ligation probe amplification and/or array comparative genomic hybridization. A retrospective review of clinical data was also conducted. RESULTS Most cases had morphological abnormalities in the renal-urinary tract system. Diabetes developed in 12 cases (38.7%). Hyperuricemia and hypomagnesemia were associated with six (19.3%) and 13 cases (41.9%), respectively. Pancreatic malformations were detected in seven cases (22.6%). Ten patients (32.3%) had liver abnormalities. Estimated glomerular filtration rates were significantly lower in the patients with heterozygous variants compared to those in patients harboring the deletion (median 37.6 vs 58.8 ml/min/1.73 m2; p = 0.0091). CONCLUSION We present the clinical characteristics of HNF1B-related disorders. To predict renal prognosis and complications, accurate genetic diagnosis is important. Genetic testing for HNF1B mutations should be considered for patients with renal malformations, especially when associated with other organ involvement.
Collapse
Affiliation(s)
- China Nagano
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Naoya Morisada
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan. .,Department of Clinical Genetics, Hyogo Prefectural Kobe Children's Hospital, 1-6-7, Minatojimaminami-machi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan.
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Koichi Kamei
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan
| | - Ryojiro Tanaka
- Department of Nephrology, Hyogo Prefectural Kobe Children's Hospital, 1-6-7 Minatojima Minamimachi, Chuo-ku, Kobe, Hyogo, 650-0047, Japan
| | - Shoichiro Kanda
- Department of Pediatrics, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Shinichi Shiona
- Department of Pediatrics, Oita Prefectural Hospital, 476, Oaza-Bujyo, Oita, Oita, 870-8511, Japan
| | - Yoshinori Araki
- Department of Pediatrics, Hokkaido Medical Center, 5-7-1-1 Yamanote Nishi-ku, Sapporo, Hokkaido, 063-0005, Japan
| | - Shinichiro Ohara
- Department of Pediatrics, Fukushima Medical University School of Medicine, 1 Hikarigaoka, Fukushima-shi, Fukushima, 960-1295, Japan
| | - Chieko Matsumura
- Department of Pediatrics, National Hospital Organization Chibahigashi National Hospital, 673 Nitonacho, Chuo-ku, Chiba, Chiba, 260-8712, Japan
| | - Katsuaki Kasahara
- Department of Pediatric Nephrology, Japanese Red Cross Nagoya Daini Hospital, 2-9 Myokencho, Syowa-ku, Nagoya, 4668-650, Japan
| | - Yukiko Mori
- Department of Pediatrics, Japanese Red Cross Fukui Hospital, 2-4-1, Tsukimi, Fukui, 918-8501, Japan
| | - Akane Seo
- Department of Diabetes and Endocrinology, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women's Medical University, School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Miki Washiyama
- Department of Diabetes and Endocrinology, Kusatsu General Hospital, 1660 Yabase-cho, Kusatsu, Siga, 525-8585, Japan
| | - Keisuke Sugimoto
- Department of Pediatrics, Faculty of Medicine, Kindai University, 377-2, Ohno-Higashi, Osakasayama, Osaka, 589-8511, Japan
| | - Ryoko Harada
- Department of Nephrology, Tokyo Metropolitan Children's Medical Center, 2-8-29, Musashidai, Fuchu, Tokyo, 183-8561, Japan
| | - Satoshi Tazoe
- Department of Metabolism, Osaka City General Hospital, 2-13-22 Miyakojimahondori, Miyakojima-ku, Osaka, 534-0021, Japan
| | - Hiroyo Kourakata
- Department of Respiratory Medicine, Niigata Saiseikai Sanjo Hospital, 6-18 Oonohata, Sanjyo-shi, Niigata, 955-8511, Japan
| | - Mayumi Enseki
- Department of Pediatrics, Tokai University Hospital, 143, Shimokasuya, Isehara-shi, Tokyo, 259-1193, Japan
| | - Daisuke Aotani
- Department of Gastroenterology and Metabolism, Graduate School of Medical Sciences and Medical School, Nagoya City University, 1 Kawasumi, Mizuho-cho, Mizuho-ku, Nagoya, 467-8601, Japan
| | - Takeshi Yamada
- Department of Pediatrics, Niigata University School of Medicine, 1-757, Asahimachi-dori, Chuo-ku, Niigata, 951-8510, Japan
| | - Nana Sakakibara
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Tomohiko Yamamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Shogo Minamikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| | - Kenji Ishikura
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo, 157-8535, Japan.,Department of Pediatrics, Kitasato University School of Medicine, 1-15-1, Kitasato, Minami-ku, Sagamihara, 252-0375, Japan
| | - Shuichi Ito
- Department of Pediatrics, Yokohama City University, 3-9, Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan
| | - Motoshi Hattori
- Department of Pediatric Nephrology, Tokyo Women's Medical University, School of Medicine, 8-1, Kawada-cho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | - Kazumoto Iijima
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, Hyogo, 650-0017, Japan
| |
Collapse
|
78
|
Dotto RP, de Santana LS, Lindsey SC, Caetano LA, Franco LF, Moisés RCMS, Sa JR, Nishiura JL, Teles MG, Heilberg IP, Dias-da-Silva MR, Giuffrida FMA, Reis AF. Searching for mutations in the HNF1B gene in a Brazilian cohort with renal cysts and hyperglycemia. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:250-257. [PMID: 31066763 PMCID: PMC10522195 DOI: 10.20945/2359-3997000000138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 02/13/2019] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To verify the presence of variants in HNF1B in a sample of the Brazilian population selected according to the presence of renal cysts associated with hyperglycemia. SUBJECTS AND METHODS We evaluated 28 unrelated patients with clinical suspicion of HNF1B mutation because of the concomitant presence of diabetes mellitus (DM) or prediabetes and renal cysts. Genotyping was accomplished using Sanger sequencing or multiplex ligation-dependent probe amplification (MLPA). In positive cases, available relatives were recruited. RESULTS We found two patients with HNF1B mutations. The first presented the variant p.Pro328Leufs*48(c.983delC) and had DM, renal cysts, and hypomagnesemia. The second presented a heterozygous whole gene deletion in HNF1B, DM, renal cysts, body and tail pancreatic agenesis, and hypomagnesemia; this alteration was also found in his two siblings and his father. CONCLUSION The recruitment of suspected cases of HNF1B gene mutations in Brazilians due to hyperglycemia and renal cysts presents two positive cases. Our cases contribute to the annotation of clinical and biochemical phenotypes of this rare form of maturity-onset diabetes of the young (MODY).
Collapse
Affiliation(s)
- Renata P. Dotto
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Lucas Santos de Santana
- Universidade de São PauloUnidade de Endocrinologia Genética/LIM25Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilGrupo de Diabetes Monogênico, Unidade de Endocrinologia Genética/LIM25, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil
| | - Susan C. Lindsey
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Lilian Araújo Caetano
- Universidade de São PauloUnidade de Endocrinologia Genética/LIM25Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilGrupo de Diabetes Monogênico, Unidade de Endocrinologia Genética/LIM25, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil
| | - Luciana F. Franco
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Regina Célia M. S. Moisés
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - João R. Sa
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - José Luiz Nishiura
- Universidade Federal de São PauloDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Milena Gurgel Teles
- Universidade de São PauloUnidade de Endocrinologia Genética/LIM25Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilGrupo de Diabetes Monogênico, Unidade de Endocrinologia Genética/LIM25, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil
| | - Ita P. Heilberg
- Universidade Federal de São PauloDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Magnus R. Dias-da-Silva
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Fernando M. A. Giuffrida
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
- Universidade do Estado da BahiaDepartamento de Ciências da VidaUniversidade do Estado da BahiaSalvadorBABrasilDepartamento de Ciências da Vida, Universidade do Estado da Bahia (UNEB), Salvador, BA, Brasil
| | - André F. Reis
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| |
Collapse
|
79
|
Shuster S, Keunen J, Shannon P, Watkins N, Chong K, Chitayat D. Prenatal detection of isolated bilateral hyperechogenic kidneys: Etiologies and outcomes. Prenat Diagn 2019; 39:693-700. [PMID: 30650191 DOI: 10.1002/pd.5418] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 12/25/2018] [Accepted: 12/29/2018] [Indexed: 12/27/2022]
Abstract
OBJECTIVES To delineate the etiology and outcome of prenatally diagnosed isolated bilateral hyperechogenic kidneys (IBHK). STUDY DESIGN Pregnancies with IBHK on prenatal ultrasound identified and followed by us between January 1, 2000 and January 1, 2015 were evaluated regarding the etiology and outcome by evaluation of family history, targeted AR-PKD and AD-PKD DNA analysis, and microarray analysis, according to renal size and amniotic fluid volume. RESULTS Of the 52 identified cases, there were 34 cases with enlarged kidneys, 16 with normal size kidneys, and two with small kidneys. There were seven cases with AD-PKD, six inherited, and one with de novo causative variants in the PKD1 gene. Fifteen had AR-PKD, and microarray analysis showed two inherited findings: one with 17q12 deletion including the HNF1B/TCF2 gene inherited from asymptomatic mother and a duplication at 3p26.1 inherited from a healthy father. Of the remaining four cases, three cases had bilateral multicystic dysplastic kidneys, and one had unilateral renal agenesis. CONCLUSION Microarray analysis and mutation analysis for PKD1 and PKHD1 have an important contribution to the diagnostic investigation of IBHK and to the management of affected and future pregnancies. Poor outcome was associated with large hyperechoic kidneys with oligohydramnios.
Collapse
Affiliation(s)
- Shirley Shuster
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Johannes Keunen
- The Fetal Medicine Unit, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Patrick Shannon
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Nicholas Watkins
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Karen Chong
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.,Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - David Chitayat
- The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.,Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
80
|
Abstract
In addition to the common types of diabetes mellitus, two major monogenic diabetes forms exist. Maturity-onset diabetes of the young (MODY) represents a heterogenous group of monogenic, autosomal dominant diseases. MODY accounts for 1-2% of all diabetes cases, and it is not just underdiagnosed but often misdiagnosed to type 1 or type 2 diabetes. More than a dozen MODY genes have been identified to date, and their molecular classification is of great importance in the correct treatment decision and in the judgment of the prognosis. The most prevalent subtypes are HNF1A, GCK, and HNF4A. Genetic testing for MODY has changed recently due to the technological advancements, as contrary to the sequential testing performed in the past, nowadays all MODY genes can be tested simultaneously by next-generation sequencing. The other major group of monogenic diabetes is neonatal diabetes mellitus which can be transient or permanent, and often the diabetes is a part of a syndrome. It is a severe monogenic disease appearing in the first 6 months of life. The hyperglycemia usually requires insulin. There are two forms, permanent neonatal diabetes mellitus (PNDM) and transient neonatal diabetes mellitus (TNDM). In TNDM, the diabetes usually reverts within several months but might relapse later in life. The incidence of NDM is 1:100,000-1:400,000 live births, and PNDM accounts for half of the cases. Most commonly, neonatal diabetes is caused by mutations in KCNJ11 and ABCC8 genes encoding the ATP-dependent potassium channel of the β cell. Neonatal diabetes has experienced a quick and successful transition into the clinical practice since the discovery of the molecular background. In case of both genetic diabetes groups, recent guidelines recommend genetic testing.
Collapse
Affiliation(s)
- Zsolt Gaál
- 4th Department of Medicine, Jósa András Teaching Hospital, Nyíregyháza, Hungary
| | - István Balogh
- Division of Clinical Genetics, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
81
|
Torell F, Bennett K, Cereghini S, Fabre M, Rännar S, Lundstedt-Enkel K, Moritz T, Haumaitre C, Trygg J, Lundstedt T. Metabolic Profiling of Multiorgan Samples: Evaluation of MODY5/RCAD Mutant Mice. J Proteome Res 2018; 17:2293-2306. [PMID: 29873499 DOI: 10.1021/acs.jproteome.7b00821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In the present study, we performed a metabolomics analysis to evaluate a MODY5/RCAD mouse mutant line as a potential model for HNF1B-associated diseases. Gas chromatography time-of-flight mass spectrometry (GC-TOF-MS) of gut, kidney, liver, muscle, pancreas, and plasma samples uncovered the tissue specific metabolite distribution. Orthogonal projections to latent structures discriminant analysis (OPLS-DA) was used to identify the differences between MODY5/RCAD and wild-type mice in each of the tissues. The differences included, for example, increased levels of amino acids in the kidneys and reduced levels of fatty acids in the muscles of the MODY5/RCAD mice. Interestingly, campesterol was found in higher concentrations in the MODY5/RCAD mice, with a four-fold and three-fold increase in kidneys and pancreas, respectively. As expected, the MODY5/RCAD mice displayed signs of impaired renal function in addition to disturbed liver lipid metabolism, with increased lipid and fatty acid accumulation in the liver. From a metabolomics perspective, the MODY5/RCAD model was proven to display a metabolic pattern similar to what would be suspected in HNF1B-associated diseases. These findings were in line with the presumed outcome of the mutation based on the different anatomy and function of the tissues as well as the effect of the mutation on development.
Collapse
Affiliation(s)
- Frida Torell
- Computational Life Science Cluster (CLiC), Department of Chemistry , Umeå University , Umeå 90187 , Sweden.,Accelerator Lab (ACL) , Karlsruhe Institute of Technology , Karlsruhe 76344 , Germany
| | | | - Silvia Cereghini
- CNRS, UMR7622, 75005 Paris , France.,UPMC, UMR7622 , Sorbonne Universites , 75005 Paris , France.,Inserm U-1156 Paris , France
| | - Mélanie Fabre
- CNRS, UMR7622, 75005 Paris , France.,UPMC, UMR7622 , Sorbonne Universites , 75005 Paris , France.,Inserm U-1156 Paris , France
| | | | - Katrin Lundstedt-Enkel
- AcureOmics AB, Umeå 90736 , Sweden.,Department of Organismal Biology , Uppsala University , Uppsala 75236 , Sweden
| | - Thomas Moritz
- AcureOmics AB, Umeå 90736 , Sweden.,Umeå Plant Science Centre, Department of Forest Genetics and Plant Physiology , Swedish University of Agricultural Sciences , Umeå 901 87 , Sweden
| | - Cécile Haumaitre
- CNRS, UMR7622, 75005 Paris , France.,UPMC, UMR7622 , Sorbonne Universites , 75005 Paris , France.,Inserm U-1156 Paris , France
| | - Johan Trygg
- Computational Life Science Cluster (CLiC), Department of Chemistry , Umeå University , Umeå 90187 , Sweden
| | - Torbjörn Lundstedt
- AcureOmics AB, Umeå 90736 , Sweden.,Department of Organic Pharmaceutical Chemistry , Uppsala University , Uppsala 75123 , Sweden
| |
Collapse
|
82
|
Abstract
The liver and kidneys are often similarly affected by a single disease. This is the case in metabolic, immunological, toxic, and infectious diseases, and in the different congenital malformation syndromes. Also, an enzymatic defect in an otherwise healthy liver or the consequences of advanced liver disease by itself can cause kidney disease as a secondary phenomenon. In this review, we describe numerous pathogenic mechanisms leading to dysfunction or malformations of the liver and kidneys in children. We encourage multidisciplinary management for optimal care. A combined liver-kidney transplantation is sometimes needed.
Collapse
|
83
|
Pace NP, Craus J, Felice A, Vassallo J. Case Report: Identification of an HNF1B p.Arg527Gln mutation in a Maltese patient with atypical early onset diabetes and diabetic nephropathy. BMC Endocr Disord 2018; 18:28. [PMID: 29764441 PMCID: PMC5952643 DOI: 10.1186/s12902-018-0257-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 05/03/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The diagnosis of atypical non-autoimmune forms of diabetes mellitus, such as maturity onset diabetes of the young (MODY) presents several challenges, in view of the extensive clinical and genetic heterogeneity of the disease. In this report we describe a case of atypical non autoimmune diabetes associated with a damaging HNF1β mutation. This is distinguished by a number of uncharacteristic clinical features, including early-onset obesity, the absence of renal cysts and diabetic nephropathy. HNF1β-MODY (MODY5) is an uncommon form of monogenic diabetes that is often complicated by a wide array of congenital morphological anomalies of the urinary tract, including renal cysts. This report expands on the clinical phenotypes that have been described in the context of HNF1β mutations, and is relevant as only isolated cases of diabetic nephropathy in the setting of MODY5 have been reported. CASE PRESENTATION An obese Maltese female with non-autoimmune diabetes, microalbuminuria, glomerular hyperfiltration, fatty liver and no renal cysts was studied by whole exome sequencing to investigate potential genes responsible for the proband's phenotype. A rare missense mutation at a highly conserved site in exon 8 of HNF1β was identified (c.1580G > A, NM_000458.3, p.Arg527Gln), with multiple in-silico predictions consistent with pathogenicity. This mutation has not been previously characterised. Additionally, several common susceptibility variants associated with early-onset obesity, polygenic type 2 diabetes and nephropathy were identified in the proband that could impose additional effects on the phenotype, its severity or its clinical course. CONCLUSION This report highlights several atypical features in a proband with atypical diabetes associated with an HNF1β missense mutation. It also reinforces the concept that monogenic causes of diabetes could be significant contributors to disease burden in obese individuals with atypical diabetes.
Collapse
Affiliation(s)
- Nikolai Paul Pace
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Johann Craus
- Department of Obstetrics and Gynaecology, University of Malta, Msida, Malta
| | - Alex Felice
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | | |
Collapse
|
84
|
Stiles CE, Thuraisingham R, Bockenhauer D, Platts L, Kumar AV, Korbonits M. De novo HNF1 homeobox B mutation as a cause for chronic, treatment-resistant hypomagnesaemia. Endocrinol Diabetes Metab Case Rep 2018; 2018:EDM170120. [PMID: 29576871 PMCID: PMC5863246 DOI: 10.1530/edm-17-0120] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 02/27/2018] [Indexed: 12/23/2022] Open
Abstract
29-year-old female presenting with an 8-year history of unexplained hypomagnesaemia, which was severe enough to warrant intermittent inpatient admission for intravenous magnesium. Urinary magnesium was inappropriately normal in the context of hypomagnesaemia indicating magnesium wasting. Ultrasound imaging demonstrated unilateral renal cysts and computed tomography of kidneys, ureters and bladder showed a bicornuate uterus. Referral to genetic services and subsequent testing revealed a de novo HNF1B deletion.
Collapse
Affiliation(s)
- C E Stiles
- Department of EndocrinologyWilliam Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | | | - D Bockenhauer
- UCL centre for Nephrology and Great Ormond Street Hospital NHS TrustLondon, UK
| | - L Platts
- North East Thames Regional Genetics LaboratoryGreat Ormond Street Hospital NHS Trust, London, UK
| | - A V Kumar
- North East Thames Regional Genetics ServiceGreat Ormond Street Hospital NHS Trust, London, UK
| | - M Korbonits
- Department of EndocrinologyWilliam Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
85
|
Ashton EJ, Legrand A, Benoit V, Roncelin I, Venisse A, Zennaro MC, Jeunemaitre X, Iancu D, Van't Hoff WG, Walsh SB, Godefroid N, Rotthier A, Del Favero J, Devuyst O, Schaefer F, Jenkins LA, Kleta R, Dahan K, Vargas-Poussou R, Bockenhauer D. Simultaneous sequencing of 37 genes identified causative mutations in the majority of children with renal tubulopathies. Kidney Int 2018; 93:961-967. [PMID: 29398133 DOI: 10.1016/j.kint.2017.10.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 09/29/2017] [Accepted: 10/05/2017] [Indexed: 12/25/2022]
Abstract
The clinical diagnosis of inherited renal tubulopathies can be challenging as they are rare and characterized by significant phenotypic variability. Advances in sequencing technologies facilitate the establishment of a molecular diagnosis. Therefore, we determined the diagnostic yield of a next generation sequencing panel assessing relevant disease genes in children followed through three national networks with a clinical diagnosis of a renal tubulopathy. DNA was amplified with a kit provided by the European Consortium for High-Throughput Research in Rare Kidney Diseases with nine multiplex PCR reactions. This kit produced 571 amplicons covering 37 genes associated with tubulopathies followed by massive parallel sequencing and bioinformatic interpretation. Identified mutations were confirmed by Sanger sequencing. Overall, 384 index patients and 16 siblings were assessed. Most common clinical diagnoses were 174 patients with Bartter/Gitelman syndrome and 76 with distal renal tubular acidosis. A total of 269 different variants were identified in 27 genes, of which 95 variants were considered likely, 136 definitely pathogenic and 100 had not been described at annotation. These mutations established a genetic diagnosis in 245 of the index patients. Genetic testing changed the clinical diagnosis in 16 cases and provided insights into the phenotypic spectrum of the respective disorders. Our results demonstrate a high diagnostic yield of genetic testing in children with a clinical diagnosis of a renal tubulopathy, consistent with a predominantly genetic etiology in known disease genes. Thus, genetic testing helped establish a definitive diagnosis in almost two-thirds of patients thereby informing prognosis, management and genetic counseling.
Collapse
Affiliation(s)
- Emma J Ashton
- North East Thames Regional Genetics Service Laboratories, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, UK
| | - Anne Legrand
- Department of Genetics, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France; Faculty of Medicine, Paris Sorbonne Cité, Université Paris Descartes, Paris, France
| | - Valerie Benoit
- Center of Human Genetics, Institut de Pathologie et Génétique, Gosselies, Belgium
| | - Isabelle Roncelin
- Department of Genetics, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Annabelle Venisse
- Department of Genetics, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
| | - Maria-Christina Zennaro
- Department of Genetics, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France; Faculty of Medicine, Paris Sorbonne Cité, Université Paris Descartes, Paris, France; Institut National de la Santé et la Recherche Médicale, Unité Mixte de Recherche en Santé 970, Paris-Cardiovascular Research Center, Paris, France
| | - Xavier Jeunemaitre
- Department of Genetics, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France; Faculty of Medicine, Paris Sorbonne Cité, Université Paris Descartes, Paris, France; Institut National de la Santé et la Recherche Médicale, Unité Mixte de Recherche en Santé 970, Paris-Cardiovascular Research Center, Paris, France
| | - Daniela Iancu
- Centre for Nephrology, University College London, London, UK
| | - William G Van't Hoff
- Department of Pediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Stephen B Walsh
- Centre for Nephrology, University College London, London, UK
| | - Nathalie Godefroid
- Division of Nephrology, Université Catholique de Louvain Medical School, Brussels, Belgium
| | | | | | - Olivier Devuyst
- Institute of Physiology, Zurich Center for Integrative Human Physiology, Mechanisms of Inherited Kidney Disorders Group, University of Zurich, Zurich, Switzerland; Division of Nephrology, Université Catholique de Louvain Medical School, Brussels, Belgium
| | - Franz Schaefer
- Division of Paediatric Nephrology, Heidelberg University Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | - Lucy A Jenkins
- North East Thames Regional Genetics Service Laboratories, Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, UK
| | - Robert Kleta
- Centre for Nephrology, University College London, London, UK; Department of Pediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Karin Dahan
- Center of Human Genetics, Institut de Pathologie et Génétique, Gosselies, Belgium; Division of Nephrology, Université Catholique de Louvain Medical School, Brussels, Belgium
| | - Rosa Vargas-Poussou
- Department of Genetics, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France; Faculty of Medicine, Paris Sorbonne Cité, Université Paris Descartes, Paris, France.
| | - Detlef Bockenhauer
- Centre for Nephrology, University College London, London, UK; Department of Pediatric Nephrology, Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
86
|
König JC, Titieni A, Konrad M. Network for Early Onset Cystic Kidney Diseases-A Comprehensive Multidisciplinary Approach to Hereditary Cystic Kidney Diseases in Childhood. Front Pediatr 2018; 6:24. [PMID: 29497606 PMCID: PMC5819567 DOI: 10.3389/fped.2018.00024] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/25/2018] [Indexed: 12/16/2022] Open
Abstract
Hereditary cystic kidney diseases comprise a complex group of genetic disorders representing one of the most common causes of end-stage renal failure in childhood. The main representatives are autosomal recessive polycystic kidney disease, nephronophthisis, Bardet-Biedl syndrome, and hepatocyte nuclear factor-1beta nephropathy. Within the last years, genetic efforts have brought tremendous progress for the molecular understanding of hereditary cystic kidney diseases identifying more than 70 genes. Yet, genetic heterogeneity, phenotypic variability, a lack of reliable genotype-phenotype correlations and the absence of disease-specific biomarkers remain major challenges for physicians treating children with cystic kidney diseases. To tackle these challenges comprehensive scientific approaches are urgently needed that match the ongoing "revolution" in genetics and molecular biology with an improved efficacy of clinical data collection. Network for early onset cystic kidney diseases (NEOCYST) is a multidisciplinary, multicenter collaborative combining a detailed collection of clinical data with translational scientific approaches addressing the genetic, molecular, and functional background of hereditary cystic kidney diseases. Consisting of seven work packages, including an international registry as well as a biobank, NEOCYST is not only dedicated to current scientific questions, but also provides a platform for longitudinal clinical surveillance and provides precious sources for high-quality research projects and future clinical trials. Funded by the German Federal Government, the NEOCYST collaborative started in February 2016. Here, we would like to introduce the rationale, design, and objectives of the network followed by a short overview on the current state of progress.
Collapse
Affiliation(s)
- Jens Christian König
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Andrea Titieni
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Martin Konrad
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | | |
Collapse
|
87
|
Bártů M, Dundr P, Němejcová K, Tichá I, Hojný H, Hájková N. The Role of HNF1B in Tumorigenesis of Solid Tumours: a Review of Current Knowledge. Folia Biol (Praha) 2018; 64:71-83. [PMID: 30394265 DOI: 10.14712/fb2018064030071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Hepatocyte nuclear factor 1-β is a transcription factor which plays a crucial role during ontogenesis in the differentiation of visceral endoderm from primitive endoderm, and is especially important for the normal development of the kidney, urogenital tract, gastrointestinal tract, liver, and pancreas. Despite the growing knowledge about the potential involvement of hepatocyte nuclear factor 1-β in the process of carcinogenesis, the exact underlying mechanism that would explain its rather varied effects in different tumours has not been sufficiently investigated. Most of the data regarding the significance of hepatocyte nuclear factor 1-β arise from genome- wide association studies and is concerned with the influence of single-nucleotide polymorphisms of hepatocyte nuclear factor 1-β on either the increased or decreased susceptibility to certain types of cancer. However, the influence of both the germinal and somatic mutations of this gene on the process of carcinogenesis is still poorly understood. According to current data, in some tumours hepatocyte nuclear factor 1-β acts as a protooncogene, while in others as a tumour suppressor gene, although the reasons for this are not clear. The exact incidence of hepatocyte nuclear factor 1-β mutations and the spectrum of tumours in which they may play a role in the process of carcinogenesis remain unknown. From the practical point of view, immunohistochemical expression of hepatocyte nuclear factor 1-β can be used in differential diagnostics of certain tumours, especially clear cell carcinoma. In our article we review the current knowledge regarding the significance of hepatocyte nuclear factor 1-β in carcinogenesis.
Collapse
Affiliation(s)
- M Bártů
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - P Dundr
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - K Němejcová
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - I Tichá
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - H Hojný
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| | - N Hájková
- Institute of Pathology, First Faculty of Medicine, Charles University and General University Hospital in Prague, Czech Republic
| |
Collapse
|
88
|
Lemoine S, Cochat P, Bertholet-Thomas A, Levi C, Bonnefoy C, Sellier-Leclerc AL, Bacchetta J. Néphrologie pédiatrique : que doit savoir un néphrologue d’adulte sur ces pathologies ? Nephrol Ther 2017; 13:495-504. [DOI: 10.1016/j.nephro.2017.01.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 01/31/2017] [Accepted: 01/31/2017] [Indexed: 11/25/2022]
|
89
|
|
90
|
Viering DHHM, de Baaij JHF, Walsh SB, Kleta R, Bockenhauer D. Genetic causes of hypomagnesemia, a clinical overview. Pediatr Nephrol 2017; 32:1123-1135. [PMID: 27234911 PMCID: PMC5440500 DOI: 10.1007/s00467-016-3416-3] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 05/02/2016] [Accepted: 05/04/2016] [Indexed: 12/16/2022]
Abstract
Magnesium is essential to the proper functioning of numerous cellular processes. Magnesium ion (Mg2+) deficits, as reflected in hypomagnesemia, can cause neuromuscular irritability, seizures and cardiac arrhythmias. With normal Mg2+ intake, homeostasis is maintained primarily through the regulated reabsorption of Mg2+ by the thick ascending limb of Henle's loop and distal convoluted tubule of the kidney. Inadequate reabsorption results in renal Mg2+ wasting, as evidenced by an inappropriately high fractional Mg2+ excretion. Familial renal Mg2+ wasting is suggestive of a genetic cause, and subsequent studies in these hypomagnesemic families have revealed over a dozen genes directly or indirectly involved in Mg2+ transport. Those can be classified into four groups: hypercalciuric hypomagnesemias (encompassing mutations in CLDN16, CLDN19, CASR, CLCNKB), Gitelman-like hypomagnesemias (CLCNKB, SLC12A3, BSND, KCNJ10, FYXD2, HNF1B, PCBD1), mitochondrial hypomagnesemias (SARS2, MT-TI, Kearns-Sayre syndrome) and other hypomagnesemias (TRPM6, CNMM2, EGF, EGFR, KCNA1, FAM111A). Although identification of these genes has not yet changed treatment, which remains Mg2+ supplementation, it has contributed enormously to our understanding of Mg2+ transport and renal function. In this review, we discuss general mechanisms and symptoms of genetic causes of hypomagnesemia as well as the specific molecular mechanisms and clinical phenotypes associated with each syndrome.
Collapse
Affiliation(s)
- Daan H H M Viering
- Centre for Nephrology, University College London, London, UK
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Stephen B Walsh
- Centre for Nephrology, University College London, London, UK
| | - Robert Kleta
- Centre for Nephrology, University College London, London, UK.
- Paediatric Nephrology, Great Ormond Street Hospital, London, UK.
| | - Detlef Bockenhauer
- Centre for Nephrology, University College London, London, UK
- Paediatric Nephrology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
91
|
Wang Y, Zhao Y, Zhang J, Yang Y, Liu F. A case of a novel mutation in HNF1β-related maturity-onset diabetes of the young type 5 with diabetic kidney disease complication in a Chinese family. J Diabetes Complications 2017; 31:1243-1246. [PMID: 28502589 DOI: 10.1016/j.jdiacomp.2016.11.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Revised: 11/08/2016] [Accepted: 11/09/2016] [Indexed: 02/05/2023]
Abstract
AIMS Precise diagnosis of maturity-onset diabetes of the young (MODY) has proven valuable for understanding mechanism of diabetes and selecting optimal therapy. A proband and her mother with diabetic kidney disease (DKD) were studied to investigate potential genes responsible for diabetes and different severity of DKD between the parent and offspring. METHODS The family with suspected MODY underwent mutational analyses by the whole exome sequencing (WES). Candidate pathogenic variants were validated by Sanger sequencing and tested for co-segregation. The clinical parameters of subjects were collected from medical records. RESULTS A novel missense heterozygous mutation in exon 4 of the hepatocyte nuclear factor 1β (HNF1β), c.1007A > G (p.H336R), was identified in both the proband and her mother. Moreover, comparing the family's WES results, we found that the proband had acquired a KCNQ1 gene mutation from her father and acquired ACE and SORBS1 gene mutations from her mother. These three genes are known susceptibility genes of DKD and may impose additional effects contributing to DKD severity. CONCLUSIONS A novel mutation in HNF1β-MODY was identified in a Chinese family complicated with DKD, and the additional effect of pathogenic variants in susceptibility genes was speculated to contribute to DKD severity.
Collapse
Affiliation(s)
- Yiting Wang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yingwang Zhao
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Junlin Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuxiang Yang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.
| |
Collapse
|
92
|
Zhang R, Marsch F, Kause F, Degenhardt F, Schmiedeke E, Märzheuser S, Hoppe B, Bachour H, Boemers TM, Schäfer M, Spychalski N, Neser J, Leonhardt J, Kosch F, Ure B, Gómez B, Lacher M, Deffaa OJ, Palta M, Wittekindt B, Kleine K, Schmedding A, Grasshoff-Derr S, Ven AVD, Heilmann-Heimbach S, Zwink N, Jenetzky E, Ludwig M, Reutter H. Array-based molecular karyotyping in 115 VATER/VACTERL and VATER/VACTERL-like patients identifies disease-causing copy number variations. Birth Defects Res 2017; 109:1063-1069. [DOI: 10.1002/bdr2.1042] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/13/2017] [Accepted: 04/06/2017] [Indexed: 12/13/2022]
Affiliation(s)
- Rong Zhang
- Institute of Human Genetics; University of Bonn; Bonn Germany
- Department of Genomics; Life & Brain Center; Bonn Germany
| | - Florian Marsch
- Institute of Human Genetics; University of Bonn; Bonn Germany
| | - Franziska Kause
- Institute of Human Genetics; University of Bonn; Bonn Germany
| | - Franziska Degenhardt
- Institute of Human Genetics; University of Bonn; Bonn Germany
- Department of Genomics; Life & Brain Center; Bonn Germany
| | - Eeberhard Schmiedeke
- Department of Pediatric Surgery and Urology, Centre for Child and Youth Health; Klinikum Bremen-Mitte; Bremen Germany
| | - Stefanie Märzheuser
- Department of Pediatric Surgery, Campus Virchow Clinic; Charité University Hospital Berlin; Berlin Germany
| | - Bernd Hoppe
- Department of Pediatrics; University Medical Center; Bonn Germany
| | - Haitham Bachour
- Department of Pediatric Surgery; University Hospital Bonn; Bonn Germany
| | - Thomas M. Boemers
- Department of Pediatric Surgery and Urology; University Hospital Cologne; Cologne Germany
| | - Matthias Schäfer
- Department of Pediatric Surgery and Urology; Cnopf'sche Kinderklinik; Nürnberg Germany
| | - Nicole Spychalski
- Department of Pediatric Surgery and Urology; Cnopf'sche Kinderklinik; Nürnberg Germany
| | - Jörg Neser
- Department of Pediatric Surgery; General Hospital; Chemnitz Germany
| | - Johannes Leonhardt
- Department of Pediatric Surgery; St. Bernward-Krankenhaus; Hildesheim Germany
| | - Ferdinand Kosch
- Department of Pediatric Surgery; Städtisches Klinikum Karlsruhe; Karlsruhe Germany
| | - Benno Ure
- Center of Pediatric Surgery Hannover; Hannover Medical School and Bult Children's Hospital; Hannover Germany
| | - Barbara Gómez
- Department of Pediatric Surgery; Children's and Youth Hospital “Auf der Bult”; Hannover Germany
| | - Martin Lacher
- Department of Pediatric Surgery; University of Leipzig; Leipzig Germany
| | - Oliver J. Deffaa
- Department of Pediatric Surgery; University of Leipzig; Leipzig Germany
| | - Markus Palta
- Department of Pediatric Surgery; Evangelisches Krankenhaus Hamm; Hamm Germany
| | - Boris Wittekindt
- Clinic for Pediatric and Adolescent Medicine; University Hospital; Frankfurt Germany
| | - Katharina Kleine
- Department of Pediatric Surgery; Evangelisches Krankenhaus Oberhausen; Germany
| | - Andrea Schmedding
- Department of Paediatric Surgery and Paediatric Urology; University Hospital of the Goethe-University Frankfurt/M; Frankfurt/M Germany
| | | | - Amelie van der Ven
- Institute of Human Genetics; University of Bonn; Bonn Germany
- Department of Medicine, Boston Children's Hospital; Harvard Medical School; Boston Massachusetts
| | - Stefanie Heilmann-Heimbach
- Institute of Human Genetics; University of Bonn; Bonn Germany
- Department of Genomics; Life & Brain Center; Bonn Germany
| | - Nadine Zwink
- Division of Clinical Epidemiology and Aging Research; German Cancer Research Center; Heidelberg Germany
| | - Ekkehart Jenetzky
- Division of Clinical Epidemiology and Aging Research; German Cancer Research Center; Heidelberg Germany
- Department for Child and Adolescent Psychiatry; Johannes Gutenberg-University; Mainz Germany
- Child Centre Maulbronn; Hospital for Paediatric Neurology and Social Paediatrics; Maulbronn Germany
| | - Michael Ludwig
- Department of Clinical Chemistry and Clinical Pharmacology; University of Bonn; Bonn Germany
| | - Heiko Reutter
- Institute of Human Genetics; University of Bonn; Bonn Germany
- Department of Genomics; Life & Brain Center; Bonn Germany
- Department of Neonatology and Pediatric Intensive Care, Children's Hospital; University of Bonn; Bonn Germany
| |
Collapse
|
93
|
Abstract
PURPOSE OF REVIEW Magnesium (Mg) imbalances are frequently overlooked. Hypermagnesemia usually occurs in preeclamptic women after Mg therapy or in end-stage renal disease patients, whereas hypomagnesemia is more common with a prevalence of up to 15% in the general population. Increasing evidence points toward a role for mild-to-moderate chronic hypomagnesemia in the pathogenesis of hypertension, type 2 diabetes mellitus, and metabolic syndrome. RECENT FINDINGS The kidneys are the major regulator of total body Mg homeostasis. Over the last decade, the identification of the responsible genes in rare genetic disorders has enhanced our understanding of how the kidney handles Mg. The different genetic disorders and medications contributing to abnormal Mg homeostasis are reviewed. SUMMARY As dysfunctional Mg homeostasis contributes to the development of many common human disorders, serum Mg deserves closer monitoring. Hypomagnesemic patients may be asymptomatic or may have mild symptoms. In severe hypomagnesemia, patients may present with neurological symptoms such as seizures, spasms, or cramps. Renal symptoms include nephrocalcinosis and impaired renal function. Most conditions affect tubular Mg reabsorption by disturbing the lumen-positive potential in the thick ascending limb or the negative membrane potential in the distal convoluted tubule.
Collapse
|
94
|
Misurac J. Chronic kidney disease in the neonate: etiologies, management, and outcomes. Semin Fetal Neonatal Med 2017; 22:98-103. [PMID: 27733241 DOI: 10.1016/j.siny.2016.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Neonatal chronic kidney disease (CKD) occurs with an estimated incidence of 1 in 10,000 live births, whereas the incidence of neonatal end-stage renal disease (ESRD) is about 7.1 per million age-related population. The most frequent etiologies are renal hypoplasia/dysplasia, posterior urethral valves, and other congenital anomalies of the kidney and urinary tract. Other etiologies include polycystic kidney disease, cortical necrosis, and renal vascular thrombosis. Management of CKD focuses primarily on replacing renal functions such as erythropoietin, 1,25-hydroxylation of vitamin D, electrolyte homeostasis/excretion, and, in ESRD, waste product removal. Nutrition and growth monitoring are of utmost importance, with the majority of ESRD infants requiring gastrostomy tube for nutrition. Outcomes of neonates (<31 days) started on dialysis continue to improve, with large cohort studies showing 2-3-year survival rates of 79-81%. As in other neonatal disciplines, the gestational age and size limits for safe provision of dialysis continue to decrease.
Collapse
Affiliation(s)
- Jason Misurac
- Pediatric Nephrology, University of Iowa Children's Hospital, Iowa City, IA, USA.
| |
Collapse
|
95
|
Abstract
Congenital abnormalities of the kidney and urinary tract (CAKUT) are one of the leading congenital defects to be identified on prenatal ultrasound. CAKUT represent a broad spectrum of abnormalities, from transient hydronephrosis to severe bilateral renal agenesis. CAKUT are a major contributor to chronic and end stage kidney disease (CKD/ESKD) in children. Prenatal imaging is useful to identify CAKUT, but will not detect all defects. Both genetic abnormalities and the fetal environment contribute to CAKUT. Monogenic gene mutations identified in human CAKUT have advanced our understanding of molecular mechanisms of renal development. Low nephron number and solitary kidneys are associated with increased risk of adult onset CKD and ESKD. Premature and low birth weight infants represent a high risk population for low nephron number. Additional research is needed to identify biomarkers and appropriate follow-up of premature and low birth weight infants into adulthood.
Collapse
Affiliation(s)
- Stacy Rosenblum
- Department of Pediatrics/Neonatology, Children's Hospital of Montefiore/Einstein, Bronx, NY, USA
| | - Abhijeet Pal
- Department of Pediatrics/Nephrology, Children's Hospital of Montefiore/Einstein, Bronx, NY, USA
| | - Kimberly Reidy
- Department of Pediatrics/Nephrology, Children's Hospital of Montefiore/Einstein, Bronx, NY, USA.
| |
Collapse
|
96
|
Pharmacogenetics of posttransplant diabetes mellitus. THE PHARMACOGENOMICS JOURNAL 2017; 17:209-221. [DOI: 10.1038/tpj.2017.1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/04/2016] [Accepted: 01/09/2017] [Indexed: 02/08/2023]
|
97
|
Genetic risk factors for ovarian cancer and their role for endometriosis risk. Gynecol Oncol 2017; 145:142-147. [PMID: 28214017 DOI: 10.1016/j.ygyno.2017.02.022] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Several genetic variants have been validated as risk factors for ovarian cancer. Endometriosis has also been described as a risk factor for ovarian cancer. Identifying genetic risk factors that are common to the two diseases might help improve our understanding of the molecular pathogenesis potentially linking the two conditions. METHODS In a hospital-based case-control analysis, 12 single nucleotide polymorphisms (SNPs), validated by the Ovarian Cancer Association Consortium (OCAC) and the Collaborative Oncological Gene-environment Study (COGS) project, were genotyped using TaqMan® OpenArray™ analysis. The cases consisted of patients with endometriosis, and the controls were healthy individuals without endometriosis. A total of 385 cases and 484 controls were analyzed. Odds ratios and P values were obtained using simple logistic regression models, as well as from multiple logistic regression models with adjustment for clinical predictors. RESULTS rs11651755 in HNF1B was found to be associated with endometriosis in this case-control study. The OR was 0.66 (95% CI, 0.51 to 0.84) and the P value after correction for multiple testing was 0.01. None of the other genotypes was associated with a risk for endometriosis. CONCLUSIONS As rs11651755 in HNF1B modified both the ovarian cancer risk and also the risk for endometriosis, HNF1B may be causally involved in the pathogenetic pathway leading from endometriosis to ovarian cancer.
Collapse
|
98
|
Thomas CP, Mansilla MA, Sompallae R, Mason SO, Nishimura CJ, Kimble MJ, Campbell CA, Kwitek AE, Darbro BW, Stewart ZA, Smith RJH. Screening of Living Kidney Donors for Genetic Diseases Using a Comprehensive Genetic Testing Strategy. Am J Transplant 2017; 17:401-410. [PMID: 27434427 PMCID: PMC5297870 DOI: 10.1111/ajt.13970] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Revised: 06/20/2016] [Accepted: 07/12/2016] [Indexed: 01/25/2023]
Abstract
Related living kidney donors (LKDs) are at higher risk of end-stage renal disease (ESRD) compared with unrelated LKDs. A genetic panel was developed to screen 115 genes associated with renal diseases. We used this panel to screen six negative controls, four transplant candidates with presumed genetic renal disease and six related LKDs. After removing common variants, pathogenicity was predicted using six algorithms to score genetic variants based on conservation and function. All variants were evaluated in the context of patient phenotype and clinical data. We identified causal variants in three of the four transplant candidates. Two patients with a family history of autosomal dominant polycystic kidney disease segregated variants in PKD1. These findings excluded genetic risk in three of four relatives accepted as potential LKDs. A third patient with an atypical history for Alport syndrome had a splice site mutation in COL4A5. This pathogenic variant was excluded in a sibling accepted as an LKD. In another patient with a strong family history of ESRD, a negative genetic screen combined with negative comparative genomic hybridization in the recipient facilitated counseling of the related donor. This genetic renal disease panel will allow rapid, efficient and cost-effective evaluation of related LKDs.
Collapse
Affiliation(s)
- C. P. Thomas
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA,Department of PediatricsCarver College of MedicineUniversity of IowaIowa CityIA,VA Medical CenterIowa CityIA
| | - M. A. Mansilla
- Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA
| | - R. Sompallae
- Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA
| | - S. O. Mason
- Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA
| | - C. J. Nishimura
- Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA
| | - M. J. Kimble
- Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA
| | - C. A. Campbell
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA,Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA
| | - A. E. Kwitek
- Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA,Department of PharmacologyCarver College of MedicineUniversity of IowaIowa CityIA
| | - B. W. Darbro
- Department of PediatricsCarver College of MedicineUniversity of IowaIowa CityIA,Interdisciplinary Program in GeneticsUniversity of IowaIowa CityIA,The Holden Comprehensive Cancer CenterUniversity of IowaIowa CityIA
| | - Z. A. Stewart
- Department of SurgeryDivision of Transplant SurgeryCarver College of MedicineUniversity of IowaIowa CityIA
| | - R. J. H. Smith
- Department of Internal MedicineCarver College of MedicineUniversity of IowaIowa CityIA,Department of PediatricsCarver College of MedicineUniversity of IowaIowa CityIA,Iowa Institute of Human GeneticsCarver College of MedicineUniversity of IowaIowa CityIA,Interdisciplinary Program in GeneticsUniversity of IowaIowa CityIA,Department of OtorhinolaryngologyCarver College of MedicineUniversity of IowaIowa CityIA
| |
Collapse
|
99
|
Duval H, Michel-Calemard L, Gonzales M, Loget P, Beneteau C, Buenerd A, Joubert M, Denis-Musquer M, Clemenson A, Chesnais AL, Blesson S, De Pinieux I, Delezoide AL, Bonyhay G, Bellanné-Chantelot C, Heidet L, Dupré F, Collardeau-Frachon S. Fetal anomalies associated withHNF1Bmutations: report of 20 autopsy cases. Prenat Diagn 2016; 36:744-51. [DOI: 10.1002/pd.4858] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 05/30/2016] [Accepted: 06/04/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Hélène Duval
- Service de Pathologie; Centre Hospitalier Princesse Grace; Avenue Pasteur Monaco
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
| | - Laurence Michel-Calemard
- Service d'Endocrinologie Moléculaire et Maladies Rares; Centre de Biologie et Pathologie Est; Bron France
| | - Marie Gonzales
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Service de Génétique et d'Embryologie Médicales, Hôpital Armand Trousseau, APHP; UPMC; Paris France
| | - Philippe Loget
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Laboratoire d'Anatomie et de Cytologie Pathologiques; Hôpital Pontchaillou; Rennes France
| | - Claire Beneteau
- Service de Génétique Médicale, Institut de Biologie; CHU de Nantes; Nantes France
| | - Annie Buenerd
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Centre de Pathologie Est, Hôpital Femme-Mère-Enfant, Groupement Hospitalier Est; Hospices Civils de Lyon; Lyon France
| | - Madeleine Joubert
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Service d'Anatomie Pathologique; CHU Hôtel Dieu; Nantes France
| | | | - Alix Clemenson
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Service d'Anatomie et Cytologie Pathologiques; CHU Saint-Etienne; Saint-Etienne France
| | - Anne-Laure Chesnais
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Laboratoire d'Anatomie Pathologique-Neuropathologique; Hôpital de la Timone; Marseille France
| | - Sophie Blesson
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Service de Génétique, Centre Olympes de Gouges, Hôpital Bretonneau; CHRU de Tours; Tours France
| | | | - Anne-Lise Delezoide
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Service de Biologie du Développement, Hôpital Robert Debré, APHP; Université Paris Diderot; Paris France
| | - Gheorghe Bonyhay
- Département de Génétique; Université Pierre et Marie Curie; Paris France
| | | | - Laurence Heidet
- Centre de Référence des Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA), AP-HP; Hôpital Necker; Paris France
| | - Florence Dupré
- Service de Pathologie; Centre Hospitalier Princesse Grace; Avenue Pasteur Monaco
| | - Sophie Collardeau-Frachon
- SOFFOET, Société Française de Fœtopathologie; Lyon, Rennes France
- Centre de Pathologie Est, Hôpital Femme-Mère-Enfant, Groupement Hospitalier Est; Hospices Civils de Lyon; Lyon France
- Université Claude Bernard Lyon 1; Lyon France
| |
Collapse
|
100
|
Abstract
A number of inherited renal diseases present with renal cysts and often lead to end-stage renal disease. With recent advances in genetics, increasing number of genes and mutations have been associated with cystic renal diseases. Although genetic testing can provide a definite diagnosis, it is often reserved for equivocal cases or for ongoing investigational research. Therefore, imaging findings are essential in the routine diagnosis, follow-up, and detection of complications in patients with inherited cystic renal diseases. In this article, the most recent classification, genetic analysis, clinical presentations, and imaging findings of inherited cystic renal diseases will be discussed.
Collapse
|