1
|
Dai Y, Li H, Fan S, Wang K, Cui Z, Zhao X, Sun X, Lin M, Li J, Gao Y, Tian Z, Yang H, Zha B, Lv L, Xu Y. Dimethyl fumarate promotes the degradation of HNF1B and suppresses the progression of clear cell renal cell carcinoma. Cell Death Dis 2025; 16:71. [PMID: 39915461 PMCID: PMC11802756 DOI: 10.1038/s41419-025-07412-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 12/19/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025]
Abstract
Clear cell renal cell carcinoma (ccRCC) is the most lethal subtype of renal cancer, and its treatment options remain limited. Therefore, there is an urgent need to discover therapeutic agents for ccRCC treatment. Here, we demonstrate that dimethyl fumarate (DMF), an approved medication for multiple sclerosis [1] and psoriasis, can inhibit the proliferation of ccRCC cells. Mechanistically, hepatocyte nuclear factor 1β (HNF1B), a transcription factor highly expressed in ccRCC, is succinated by DMF at cysteine residues, leading to its proteasomal degradation. Furthermore, HNF1B interacts with and stabilizes Yes-associated protein (YAP), thus DMF-mediated HNF1B degradation decreases YAP protein level and the expression of its target genes, resulting in the suppression of ccRCC cell proliferation. Importantly, oral administration of DMF sensitizes ccRCC to sunitinib treatment and enhances its efficacy in mice. In summary, we provide evidences supporting DMF as a potential drug for clinical treatment of ccRCC by targeting HNF1B and reveal a previously unrecognized role of HNF1B in regulating YAP in ccRCC.
Collapse
Affiliation(s)
- Yue Dai
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hongchen Li
- Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shiyin Fan
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Kai Wang
- Department of Endocrinology, Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Ziyi Cui
- Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xinyu Zhao
- Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xue Sun
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Mingen Lin
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jiaxi Li
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yi Gao
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziyin Tian
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hui Yang
- Department of Neurosurgery, Huashan Hospital, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Bingbing Zha
- Department of Endocrinology, Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China.
| | - Lei Lv
- Fifth People's Hospital of Shanghai, MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Yanping Xu
- Tongji Hospital, Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| |
Collapse
|
2
|
Devuyst O, Ahn C, Barten TR, Brosnahan G, Cadnapaphornchai MA, Chapman AB, Cornec-Le Gall E, Drenth JP, Gansevoort RT, Harris PC, Harris T, Horie S, Liebau MC, Liew M, Mallett AJ, Mei C, Mekahli D, Odland D, Ong AC, Onuchic LF, P-C Pei Y, Perrone RD, Rangan GK, Rayner B, Torra R, Mustafa R, Torres VE. KDIGO 2025 Clinical Practice Guideline for the Evaluation, Management, and Treatment of Autosomal Dominant Polycystic Kidney Disease (ADPKD). Kidney Int 2025; 107:S1-S239. [PMID: 39848759 DOI: 10.1016/j.kint.2024.07.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 07/17/2024] [Indexed: 01/25/2025]
|
3
|
König L, Schmidts M. The role of chromatin-related epigenetic modulations in CAKUT. Curr Top Dev Biol 2025; 163:169-227. [PMID: 40254345 DOI: 10.1016/bs.ctdb.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) represent a major health burden in humans. Phenotypes range from renal hypoplasia or renal agenesis, cystic renal dysplasia, duplicated or horseshoe kidneys to obstruction of the ureteropelvic junction, megaureters, duplicated ureters, urethral valves or bladder malformations. Over the past decade, next-generation sequencing has identified numerous causative genes; however, the genetic basis of most cases remains unexplained. It is assumed that environmental factors have a significant impact on the phenotype, but, overall, the pathogenesis has remained poorly understood. Interestingly however, CAKUT is a common phenotypic feature in two human syndromes, Kabuki and Koolen-de Vries syndrome, caused by dysfunction of genes encoding for KMT2D and KANSL1, both members of protein complexes playing an important role in histone modifications. In this chapter, we discuss current knowledge regarding epigenetic modulation in renal development and a putatively under-recognized role of epigenetics in CAKUT.
Collapse
Affiliation(s)
- Luise König
- Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | - Miriam Schmidts
- Center for Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; CIBSS-Center for Integrative Biological Signaling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
4
|
Dahmer-Heath M, Gerß J, Fliser D, Liebau MC, Speer T, Telgmann AK, Burgmaier K, Pennekamp P, Pape L, Schaefer F, Konrad M, König JC, NEOCYST Consortium 10. Urinary Dickkopf-3 Reflects Disease Severity and Predicts Short-Term Kidney Function Decline in Renal Ciliopathies. Kidney Int Rep 2025; 10:197-208. [PMID: 39810774 PMCID: PMC11725807 DOI: 10.1016/j.ekir.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 01/16/2025] Open
Abstract
Introduction Phenotypic heterogeneity and unpredictability of individual disease progression present enormous challenges in ultrarare renal ciliopathies. The tubular-derived glycoprotein, Dickkopf-related protein 3 (DKK3) is a promising biomarker for kidney fibrosis and prediction of kidney function decline. Here, we measured urinary DKK3 (uDKK3) levels in 195 pediatric patients with renal ciliopathy to assess its potential as a discriminative and prediction marker. Methods uDKK3 concentration was measured in 357 spot urine samples from 247 individuals, including 52 healthy age-matched controls. Disease entities comprised nephronophthisis (NPH) (n = 37), autosomal recessive polycystic kidney disease (ARPKD) (n = 61), Bardet Biedl syndrome (BBS) (n = 57), and hepatocyte nuclear factor 1 beta (HNF1B)-nephropathy (n = 40). The results were correlated with chronic kidney disease (CKD) stage and annual estimated glomerular filtration rate (eGFR) decline. Results Median uDKK3-to-creatinine ratios (uDKK3/crea) in all disease entities were significantly higher compared with healthy controls (11pg/mg uDKK3/crea, P < 0.001): NPH, 1.219 pg/mg; HNF1B, 731 pg/mg; BBS, 541 pg/mg; and ARPKD, 437 pg/mg. A significant correlation of CKD stage with uDKK3 levels was observed for all disease entities (P < 0.0001) with no other clinical parameter having a relevant impact. In our cohort, uDKK3 values >4.700 pg/mg were associated with a significantly greater annual eGFR loss independently of diagnosis and eGFR (P = 0.0029). Although we observed a trend toward lower uDKK3 levels in glomerulopathies compared to renal ciliopathies, there was no discriminative difference between individual ciliopathy entities (P = 0.2637). Conclusion In renal ciliopathies, uDKK3 is a marker to assess disease severity and estimate short-term kidney function decline.
Collapse
Affiliation(s)
- Mareike Dahmer-Heath
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Joachim Gerß
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
| | - Danilo Fliser
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/ Saar, Germany
| | - Max Christoph Liebau
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Center for Family Health and Center for Rare Disease, University Hospital Cologne and Medical Faculty, University of Cologne, Cologne, Germany
| | - Thimoteus Speer
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/ Saar, Germany
- Else Kroener Fresenius Center for Nephrological Research, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Kathrin Burgmaier
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Petra Pennekamp
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Lars Pape
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| | - Martin Konrad
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Jens Christian König
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - NEOCYST Consortium10
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
- Institute of Biostatistics and Clinical Research, University of Münster, Münster, Germany
- Department of Internal Medicine IV, Nephrology and Hypertension, Saarland University Medical Center, Homburg/ Saar, Germany
- Department of Pediatrics, University Hospital Cologne and Faculty of Medicine, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne, Center for Family Health and Center for Rare Disease, University Hospital Cologne and Medical Faculty, University of Cologne, Cologne, Germany
- Else Kroener Fresenius Center for Nephrological Research, University Hospital Frankfurt, Frankfurt, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
- Department of Pediatrics II, University Hospital of Essen, Essen, Germany
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, Heidelberg, Germany
| |
Collapse
|
5
|
Wei LQ, Song YB, Lan D, Miao XJ, Lin CY, Yang ST, Liu DH, Chi XJ. Impact of gout on colorectal cancer and its survival: a two-sample Mendelian randomization study. Discov Oncol 2024; 15:819. [PMID: 39708204 DOI: 10.1007/s12672-024-01714-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 12/17/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The relationship between gout and colorectal cancer (CRC) remains unclear, emphasizing the need for additional research to clarify the potential cumulative effect of gout on CRC development. METHODS Leveraging a single nucleotide polymorphism-based genome-wide association study, the potential causal correlation between gout and CRC was initially analyzed using Mendelian randomization (MR). Subsequently, our analysis was expanded to include an assessment of patient survival, with the aim of evaluating the potential causal correlation between gout and CRC and the impact of gout on CRC survival outcomes. RESULTS According to MR findings, a substantial relationship was observed between gout and the incidence of CRC when CRC was used as the outcome (OR = 0.954, 95% CI = 0.915-0.995). These results indicate a negative relationship between gout and the likelihood of developing CRC. In addition, when evaluating the overall survival (OS) or cancer-specific survival (CSS) of patients with CRC as outcomes, gout exhibited a significant relationship with survival. The inverse variance weighting approach demonstrated a progressive enhancement in CRC survival with the cumulative impact of gout (OS: OR = 2.000 × 10-4, 95% CI = 1.560 × 10-7-0.292; CSS: OR = 2.200 × 10-5, 95% CI = 4.660 × 10-9-0.104). CONCLUSION As gout accumulates, it exerts an inhibitory influence on CRC, indicating a potential protective effect. This study provides robust evidence that can guide the development of future clinical treatment approaches and research priorities.
Collapse
Affiliation(s)
- Li-Qiang Wei
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Yi-Bei Song
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Dong Lan
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Xue-Jing Miao
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Chun-Yu Lin
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Shu-Ting Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China
| | - Deng-He Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| | - Xiao-Jv Chi
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi Medical University, Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, 6 Shuangyong Road, Nanning, 530021, Guangxi Zhuang Autonomous Region, China.
| |
Collapse
|
6
|
Du W, Wang X, Zhang D, Chen W, Zuo X, Li P. A genotype-guided prediction model for the incidence of persistent acute kidney injury following lung transplantation. BMC Nephrol 2024; 25:458. [PMID: 39696008 PMCID: PMC11654156 DOI: 10.1186/s12882-024-03871-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND This study aimed to develop a nomogram for predicting persistent renal dysfunction in acute kidney injury (AKI) following lung transplantation (LTx). METHOD A total of 229 LTx patients were enrolled, and genotyping for 153 single nucleotide polymorphisms (SNPs) was performed. The cohort was randomly divided into training (n = 183) and validation (n = 46) sets in an 8:2 ratio. Statistically significant SNPs identified through pharmacogenomic analysis were combined with clinical factors to construct a comprehensive prediction model for persistent AKI using multivariate logistic regression analysis. Discrimination and calibration analyses were conducted to evaluate the performance of the model. Decision curve analysis was used to assess its clinical utility. Due to the small sample size, bootstrap internal sampling with 500 iterations was adopted for validation to prevent overfitting of the model. RESULTS The final nomogram comprised nine predictors, including body mass index, thrombin time, tacrolimus initial concentration, rs757210, rs1799884, rs6887695, rs1494558, rs2069762 and rs2275913. In the training set, the area under the receiver operating characteristic curve of the nomogram was 0.781 (95%CI: 0.715-0.846), while in the validation set it was 0.698 (95%CI: 0.542-0.855), indicating good model fit. As demonstrated by 500 Bootstrap internal sampling validations, the model has high discrimination and calibration. Additionally, decision curve analysis confirmed its clinical applicability. CONCLUSION This study presents a genotype-guided nomogram that can be used to assess the risk of persistent AKI following LTx and may assist in guiding personalized prevention strategies in clinical practice.
Collapse
Affiliation(s)
- Wenwen Du
- Department of Pharmacy, Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Xiaoxing Wang
- Department of Pharmacy, Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Dan Zhang
- Department of Pharmacy, Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Wenqian Chen
- Department of Pharmacy, Friendship Hospital, Chaoyang District, Beijing, 100029, China
| | - Xianbo Zuo
- Department of Dermatology, Department of Pharmacy, Friendship Hospital, Beijing, Chaoyang District, 100029, China
| | - Pengmei Li
- Department of Pharmacy, Friendship Hospital, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|
7
|
Sánchez-Cazorla E, Carrera N, García-González MÁ. HNF1B Transcription Factor: Key Regulator in Renal Physiology and Pathogenesis. Int J Mol Sci 2024; 25:10609. [PMID: 39408938 PMCID: PMC11476927 DOI: 10.3390/ijms251910609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 10/20/2024] Open
Abstract
The HNF1B gene, located on chromosome 17q12, encodes a transcription factor essential for the development of several organs. It regulates the expression of multiple genes in renal, pancreatic, hepatic, neurological, and genitourinary tissues during prenatal and postnatal development, influencing processes such as nephrogenesis, cellular polarity, tight junction formation, cilia development, ion transport in the renal tubule, and renal metabolism. Mutations that alter the function of Hnf1b deregulate those processes, leading to various pathologies characterized by both renal and extrarenal manifestations. The main renal diseases that develop are polycystic kidney disease, hypoplastic or dysplastic kidneys, structural abnormalities, Congenital Anomalies of the Kidney and Urinary Tract (CAKUT), and electrolyte imbalances such as hyperuricemia and hypomagnesemia. Extrarenal manifestations include Maturity-Onset Diabetes of the Young (MODY), hypertransaminasemia, genital and urinary tract malformations, Autism Spectrum Disorder (ASD), and other neurodevelopmental disorders. Patients with HNF1B alterations typically carry either punctual mutations or a monoallelic microdeletion in the 17q12 region. Future research on the molecular mechanisms and genotype-phenotype correlations in HNF1B-related conditions will enhance our understanding, leading to improved clinical management, genetic counseling, monitoring, and patient care.
Collapse
Affiliation(s)
- Eloísa Sánchez-Cazorla
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
| | - Noa Carrera
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
- RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| | - Miguel Ángel García-González
- Group of Genetics and Developmental Biology of Renal Disease, Laboratory of Nephrology, No. 11, Health Research Institute of Santiago de Compostela (IDIS), Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain;
- Genomic Medicine Group, Clinical University Hospital (CHUS), 15706 Santiago de Compostela, Spain
- RICORS 2040 (Kidney Disease), ISCIII, 15706 Santiago de Compostela, Spain
| |
Collapse
|
8
|
Jankowski J, Lee HK, Liu C, Wilflingseder J, Hennighausen L. Sexually dimorphic renal expression of mouse Klotho is directed by a kidney-specific distal enhancer responsive to HNF1b. Commun Biol 2024; 7:1142. [PMID: 39277686 PMCID: PMC11401919 DOI: 10.1038/s42003-024-06855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 09/06/2024] [Indexed: 09/17/2024] Open
Abstract
Transcription enhancers are genomic sequences regulating common and tissue-specific genes and their disruption can contribute to human disease development and progression. Klotho, a sexually dimorphic gene specifically expressed in kidney, is well-linked to kidney dysfunction and its deletion from the mouse genome leads to premature aging and death. However, the sexually dimorphic regulation of Klotho is not understood. Here, we characterize two candidate Klotho enhancers using H3K27ac epigenetic marks and transcription factor binding and investigate their functions, individually and combined, through CRISPR-Cas9 genome engineering. We discovered that only the distal (E1), but not the proximal (E2) candidate region constitutes a functional enhancer, with the double deletion not causing Klotho expression to further decrease. E1 activity is dependent on HNF1b transcription factor binding site within the enhancer. Further, E1 controls the sexual dimorphism of Klotho as evidenced by qPCR and RNA-seq. Despite the sharp reduction of Klotho mRNA, unlike germline Klotho knockouts, mutant mice present normal phenotype, including weight, lifespan, and serum biochemistry. Lastly, only males lacking E1 display more prominent acute, but not chronic kidney injury responses, indicating a remarkable range of potential adaptation to isolated Klotho loss, especially in female E1 knockouts, retaining renoprotection despite over 80% Klotho reduction.
Collapse
Affiliation(s)
- Jakub Jankowski
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, 20892, USA.
- , 8 Center Drive, Room 107, 20892, Bethesda, MD, USA.
| | - Hye Kyung Lee
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julia Wilflingseder
- Department of Physiology and Pathophysiology, University of Veterinary Medicine Vienna, 1210, Vienna, Austria
| | - Lothar Hennighausen
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
9
|
Han JY, Gwack J, Kim TY, Park J. A Korean Family Presenting with Renal Cysts and Maturity-Onset Diabetes of the Young Caused by a Novel In-Frame Deletion of HNF1B. Int J Mol Sci 2024; 25:9823. [PMID: 39337310 PMCID: PMC11432569 DOI: 10.3390/ijms25189823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Maturity-onset diabetes of the young (MODY; OMIM # 606391) comprises a cluster of inherited disorders within non-autoimmune diabetes mellitus (DM), typically emerging during adolescence or young adulthood. We report a novel in-frame deletion of HNF1B in a family with renal cysts and MODY, furthering our understanding of HNF1B-related phenotypes. We conducted sequential genetic testing to investigate the glucose intolerance, renal cysts, hepatic cysts, and agenesis of the dorsal pancreas observed in the proband. A comprehensive clinical exome sequencing approach using a Celemics G-Mendeliome Clinical Exome Sequencing Panel was employed. Considering the clinical manifestations observed in the proband, gene panel sequencing identified a heterozygous HNF1B variant, c.36_38delCCT/p.(Leu13del) (reference transcript ID: NM_000458.4), as the most likely cause of MODY in the proband. The patient's clinical presentation was consistent with MODY caused by the HNF1B variant, showing signs of glucose intolerance, renal cysts, hepatic cysts, and agenesis of the dorsal pancreas. Sanger sequencing confirmed the same HNF1B variant and established the paternally inherited autosomal dominant status of the heterozygous variant in the patient, as well as in his father and sister. The presence of early-onset diabetes, renal cysts, a family history of the condition, and nephropathy appearing before or after the diagnosis of diabetes mellitus (DM) suggests a diagnosis of HNF1B-MODY5. Early diagnosis is crucial for preventing complications of DM, enabling family screening, providing pre-conceptional genetic counseling, and monitoring kidney function decline.
Collapse
Affiliation(s)
- Ji Yoon Han
- Department of Pediatrics, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| | - Jin Gwack
- Department of Preventive Medicine, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea;
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Tae Yun Kim
- Department of Thoracic and Cardiovascular Surgery, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
| | - Joonhong Park
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
10
|
Gazeu A, Collardeau-Frachon S. Practical Approach to Congenital Anomalies of the Kidneys: Focus on Anomalies With Insufficient or Abnormal Nephron Development: Renal Dysplasia, Renal Hypoplasia, and Renal Tubular Dysgenesis. Pediatr Dev Pathol 2024; 27:459-493. [PMID: 39270126 DOI: 10.1177/10935266241239241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) accounts for up to 30% of antenatal congenital anomalies and is the main cause of kidney failure in children worldwide. This review focuses on practical approaches to CAKUT, particularly those with insufficient or abnormal nephron development, such as renal dysplasia, renal hypoplasia, and renal tubular dysgenesis. The review provides insights into the histological features, pathogenesis, mechanisms, etiologies, antenatal and postnatal presentation, management, and prognosis of these anomalies. Differential diagnoses are discussed as several syndromes may include CAKUT as a phenotypic component and renal dysplasia may occur in some ciliopathies, tumor predisposition syndromes, and inborn errors of metabolism. Diagnosis and genetic counseling for CAKUT are challenging, due to the extensive variability in presentation, genetic and phenotypic heterogeneity, and difficulties to assess postnatal lung and renal function on prenatal imaging. The review highlights the importance of perinatal autopsy and pathological findings in surgical specimens to establish the diagnosis and prognosis of CAKUT. The indications and the type of genetic testing are discussed. The aim is to provide essential insights into the practical approaches, diagnostic processes, and genetic considerations offering valuable guidance for pediatric and perinatal pathologists.
Collapse
Affiliation(s)
- Alexia Gazeu
- Department of pathology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, University Hospital of Lyon, Lyon Bron, France
- Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
| | - Sophie Collardeau-Frachon
- Department of pathology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, University Hospital of Lyon, Lyon Bron, France
- Université Claude Bernard Lyon 1, Faculté de Médecine Lyon Est, Lyon, France
- Société française de Fœtopathologie, Soffoet, Paris, France
| |
Collapse
|
11
|
Vourdoumpa A, Paltoglou G, Mertzanian A, Sertedaki A, Sakou II, Karanasios S, Karavanaki K, Charmandari E. Challenges in the management of patients with HNF1B MODY and multisystem manifestations: the cases of two adolescent boys. Hormones (Athens) 2024; 23:439-445. [PMID: 38980656 DOI: 10.1007/s42000-024-00580-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
INTRODUCTION Hepatocyte nuclear factor-1 beta (HNF1B) encodes a homeodomain-containing transcription factor, which is expressed early in embryogenesis and is involved in the development of multiple tissues and organs. HNF1B mutations cause complex multisystem disorders, with renal developmental disease and maturity onset diabetes of the young (HNF1B MODY), a rare cause of diabetes mellitus, being representative features. METHODS We present two adolescent boys from different socioeconomic backgrounds who were diagnosed with genetically confirmed HNF1B MODY following hospitalization for diabetic ketoacidosis in the first case and after diagnostic work-up due to impaired glucose tolerance in the second case. Multisystem manifestations, including pancreatic hypoplasia and early-onset diabetes mellitus (DM), renal cysts, hypomagnesemia, hyperuricemia, liver and biliary impairment, genital tract malformations, and primary hyperparathyroidism were also present, strongly suggesting HNF1B MODY. RESULTS The first patient was treated with subcutaneous insulin but was lost to follow-up due to social reasons. Conversely, early diagnosis in the second patient allowed the management of multisystem defects by a multidisciplinary team of experts. Moreover, manifestation of HNF1B MODY in the form of diabetic ketoacidosis was prevented and a structured diabetes training program has proven successful in regulating glycemic control, postponing the necessity for insulin treatment. CONCLUSION Early genetic work-up of patients with dysglycemia associated with a specific phenotype suggestive of HNF1B MODY is extremely important in the care of children and adolescents with diabetes since it ensures that early and optimal management is initiated, thereby preventing the onset of life-threatening diabetic ketoacidosis and other multisystem complications and/or comorbidities.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece.
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece.
| | - Anny Mertzanian
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, 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, 11527, Greece
| | - Irini-Ikbale Sakou
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - Spyridon Karanasios
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - Kyriaki Karavanaki
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, "P. & A. Kyriakou" Children's Hospital, Athens, 11527, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, "Aghia Sophia" Children's Hospital, Athens, 11527, Greece
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| |
Collapse
|
12
|
Rioux AV, Nsimba-Batomene TR, Slimani S, Bergeron NAD, Gravel MAM, Schreiber SV, Fiola MJ, Haydock L, Garneau AP, Isenring P. Navigating the multifaceted intricacies of the Na +-Cl - cotransporter, a highly regulated key effector in the control of hydromineral homeostasis. Physiol Rev 2024; 104:1147-1204. [PMID: 38329422 PMCID: PMC11381001 DOI: 10.1152/physrev.00027.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 01/01/2024] [Accepted: 02/03/2024] [Indexed: 02/09/2024] Open
Abstract
The Na+-Cl- cotransporter (NCC; SLC12A3) is a highly regulated integral membrane protein that is known to exist as three splice variants in primates. Its primary role in the kidney is to mediate the cosymport of Na+ and Cl- across the apical membrane of the distal convoluted tubule. Through this role and the involvement of other ion transport systems, NCC allows the systemic circulation to reclaim a fraction of the ultrafiltered Na+, K+, Cl-, and Mg+ loads in exchange for Ca2+ and [Formula: see text]. The physiological relevance of the Na+-Cl- cotransport mechanism in humans is illustrated by several abnormalities that result from NCC inactivation through the administration of thiazides or in the setting of hereditary disorders. The purpose of the present review is to discuss the molecular mechanisms and overall roles of Na+-Cl- cotransport as the main topics of interest. On reading the narrative proposed, one will realize that the knowledge gained in regard to these themes will continue to progress unrelentingly no matter how refined it has now become.
Collapse
Affiliation(s)
- A V Rioux
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - T R Nsimba-Batomene
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S Slimani
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - N A D Bergeron
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M A M Gravel
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - S V Schreiber
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - M J Fiola
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| | - L Haydock
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - A P Garneau
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
- Service de Néphrologie-Transplantation Rénale Adultes, Hôpital Necker-Enfants Malades, AP-HP, INSERM U1151, Université Paris Cité, Paris, France
| | - P Isenring
- Department of Medicine, Nephrology Research Group, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
13
|
Zečević K, Volčanšek Š, Katsiki N, Rizzo M, Milardović TM, Stoian AP, Banach M, Muzurović E. Maturity-onset diabetes of the young (MODY) - in search of ideal diagnostic criteria and precise treatment. Prog Cardiovasc Dis 2024; 85:14-25. [PMID: 38513726 DOI: 10.1016/j.pcad.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 03/17/2024] [Indexed: 03/23/2024]
Abstract
Maturity-onset diabetes of the young (MODY) is a spectrum of clinically heterogenous forms of monogenic diabetes mellitus characterized by autosomal dominant inheritance, onset at a young age, and absence of pancreatic islets autoimmunity. This rare form of hyperglycemia, with clinical features overlapping with type 1 and type 2 diabetes mellitus, has 14 subtypes with differences in prevalence and complications occurrence which tailor therapeutic approach. MODY phenotypes differ based on the gene involved, gene penetrance and expressivity. While MODY 2 rarely leads to diabetic complications and is easily managed with lifestyle interventions alone, more severe subtypes, such as MODY 1, 3, and 6, require an individualized treatment approach to maintain a patient's quality of life and prevention of complications. This review summarizes current evidence on the presentation, diagnosis, and management of MODY, an example of a genetic cause of hyperglycemia that calls for a precision medicine approach.
Collapse
Affiliation(s)
- Ksenija Zečević
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro
| | - Špela Volčanšek
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Center Ljubljana, Ljubljana, Slovenia; Medical Faculty Ljubljana, Ljubljana, Slovenia
| | - Niki Katsiki
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece; School of Medicine, European University Cyprus, Nicosia, Cyprus
| | - Manfredi Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy; Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Tanja Miličević Milardović
- Internal Medicine Department, Endocrinology, Diabetology, and Metabolism Division, University Hospital of Split, Split, Croatia; University of Split School of Medicine, Split, Croatia
| | - Anca Pantea Stoian
- Diabetes, Nutrition and Metabolic diseases Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Maciej Banach
- Department of Preventive Cardiology and Lipidology, Medical University of Łódź, Lodz, Poland; Department of Cardiology and Adult Congenital Heart Diseases, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland; Ciccarone Center for the Prevention of Cardiovascular Disease, Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Emir Muzurović
- Faculty of Medicine, University of Montenegro, Podgorica, Montenegro; Department of Internal Medicine, Endocrinology Section, Clinical Center of Montenegro, Podgorica, Montenegro.
| |
Collapse
|
14
|
Hokazono S, Imagawa E, Hirano D, Ikegami T, Oishi K, Konuma T. 1H, 13C and 15N backbone resonance assignments of hepatocyte nuclear factor-1-beta (HNF1β) POU S and POU HD. BIOMOLECULAR NMR ASSIGNMENTS 2024; 18:59-63. [PMID: 38451454 DOI: 10.1007/s12104-024-10168-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
Hepatocyte nuclear factor 1β (HNF1β) is a transcription factor that plays a key role in the development and function of the liver, pancreas, and kidney. HNF1β plays a key role in early vertebrate development and the morphogenesis of these organs. In humans, heterozygous mutations in the HNF1B gene can result in organ dysplasia, making it the most common cause of developmental renal diseases, including renal cysts, renal malformations, and familial hypoplastic glomerular cystic kidney disease. Pathogenic variants in the HNF1B gene are known to cause various diseases, including maturity-onset diabetes of the young and developmental renal diseases. This study presents the backbone resonance assignments of HNF1β POUS and POUHD domains, which are highly conserved domains required for the recognition of double-stranded DNA. Our data will be useful for NMR studies to verify the altered structures and functions of mutant HNF1B proteins that can induce developmental renal diseases, including renal cysts, renal malformations, and familial hypoplastic glomerular cystic kidney disease. This study will provide the structural basis for future studies to elucidate the molecular mechanisms underlying how mutations in HNF1β cause diseases.
Collapse
Affiliation(s)
- Sayaka Hokazono
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Eri Imagawa
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Daishi Hirano
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Takahisa Ikegami
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Kimihiko Oishi
- Department of Pediatrics, The Jikei University School of Medicine, Tokyo, Japan
| | - Tsuyoshi Konuma
- Graduate School of Medical Life Science, Yokohama City University, 1-7-29 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa, Japan.
| |
Collapse
|
15
|
Kołbuc M, Kołek MF, Motyka R, Bieniaś B, Habbig S, Burgmaier K, Prikhodina L, Papizh S, Tasic V, Okorn C, Szczepańska M, Kiliś-Pstrusińska K, Wasilewska A, Adamczyk P, Tkaczyk M, Pańczyk-Tomaszewska M, Miklaszewska M, Pawlaczyk K, Bukowska-Olech E, Jamsheer A, Jankauskiene A, König J, Cheong HI, Ahn YH, Kaspar S, Sikora P, Beck BB, Zaniew M. Development of a tool for predicting HNF1B mutations in children and young adults with congenital anomalies of the kidneys and urinary tract. Pediatr Nephrol 2024; 39:1847-1858. [PMID: 38196016 PMCID: PMC11026189 DOI: 10.1007/s00467-023-06262-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/12/2023] [Accepted: 12/12/2023] [Indexed: 01/11/2024]
Abstract
BACKGROUND We aimed to develop a tool for predicting HNF1B mutations in children with congenital abnormalities of the kidneys and urinary tract (CAKUT). METHODS The clinical and laboratory data from 234 children and young adults with known HNF1B mutation status were collected and analyzed retrospectively. All subjects were randomly divided into a training (70%) and a validation set (30%). A random forest model was constructed to predict HNF1B mutations. The recursive feature elimination algorithm was used for feature selection for the model, and receiver operating characteristic curve statistics was used to verify its predictive effect. RESULTS A total of 213 patients were analyzed, including HNF1B-positive (mut + , n = 109) and HNF1B-negative (mut - , n = 104) subjects. The majority of patients had mild chronic kidney disease. Kidney phenotype was similar between groups, but bilateral kidney anomalies were more frequent in the mut + group. Hypomagnesemia and hypermagnesuria were the most common abnormalities in mut + patients and were highly selective of HNF1B. Hypomagnesemia based on age-appropriate norms had a better discriminatory value than the age-independent cutoff of 0.7 mmol/l. Pancreatic anomalies were almost exclusively found in mut + patients. No subjects had hypokalemia; the mean serum potassium level was lower in the HNF1B cohort. The abovementioned, discriminative parameters were selected for the model, which showed a good performance (area under the curve: 0.85; sensitivity of 93.67%, specificity of 73.57%). A corresponding calculator was developed for use and validation. CONCLUSIONS This study developed a simple tool for predicting HNF1B mutations in children and young adults with CAKUT.
Collapse
Affiliation(s)
- Marcin Kołbuc
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland.
| | | | - Rafał Motyka
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland
| | - Beata Bieniaś
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Sandra Habbig
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Kathrin Burgmaier
- Department of Pediatrics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Deggendorf, Germany
| | - Larisa Prikhodina
- Division of Inherited & Acquired Kidney Diseases, Veltishev Research Clinical Institute for Pediatrics & Children Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Svetlana Papizh
- Division of Inherited & Acquired Kidney Diseases, Veltishev Research Clinical Institute for Pediatrics & Children Surgery, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Velibor Tasic
- Medical School Skopje, University Children's Hospital, 1000, Skopje, North Macedonia
| | - Christine Okorn
- Department of Pediatric Nephrology, University Hospital Essen, Essen, Germany
| | - Maria Szczepańska
- Department of Pediatrics, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, Katowice, Poland
| | | | - Anna Wasilewska
- Department of Pediatric Nephrology, University Hospital, Białystok, Poland
| | - Piotr Adamczyk
- Department of Pediatrics, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | - Marcin Tkaczyk
- Department of Pediatrics, Immunology and Nephrology, Polish Mother's Memorial Hospital Research Institute, Łódź, Poland
| | | | - Monika Miklaszewska
- Department of Pediatric Nephrology and Hypertension, Jagiellonian University Medical College, Kraków, Poland
| | - Krzysztof Pawlaczyk
- Department of Nephrology, Transplantology and Internal Medicine, Poznan University of Medical Sciences, Poznań, Poland
| | | | - Aleksander Jamsheer
- Department of Medical Genetics, Poznan University of Medical Sciences, Poznań, Poland
- Centers for Medical Genetics GENESIS, Poznań, Poland
| | - Augustina Jankauskiene
- Pediatric Center, Institute of Clinical Medicine, Vilnius University, Vilnius, Lithuania
| | - Jens König
- Department of General Pediatrics, University Children's Hospital Münster, Münster, Germany
| | - Hae Il Cheong
- Department of Pediatrics, Seoul Red Cross Hospital, Seoul, South Korea
| | - Yo Han Ahn
- Department of Pediatrics, Seoul National University College of Medicine, Seoul, South Korea
| | - Sophie Kaspar
- Institute of Human Genetics and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Przemysław Sikora
- Department of Pediatric Nephrology, Medical University of Lublin, Lublin, Poland
| | - Bodo B Beck
- Institute of Human Genetics and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Marcin Zaniew
- Department of Pediatrics, University of Zielona Góra, Zielona Góra, Poland.
| |
Collapse
|
16
|
Petrakis I, Sfakiotaki M, Bitsori M, Drosataki E, Dermitzaki K, Pleros C, Androvitsanea A, Samonakis D, Sertedaki A, Xekouki P, Galanakis E, Stylianou K. The Phenotypic Variability Associated with Hepatocyte Nuclear Factor 1B Genetic Defects Poses Challenges in Both Diagnosis and Therapy. Int J Mol Sci 2024; 25:4552. [PMID: 38674137 PMCID: PMC11050681 DOI: 10.3390/ijms25084552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 04/06/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
The evolving landscape of clinical genetics is becoming increasingly relevant in the field of nephrology. HNF1B-associated renal disease presents with a diverse array of renal and extrarenal manifestations, prominently featuring cystic kidney disease and diabetes mellitus. For the genetic analyses, whole exome sequencing (WES) and multiplex ligation-dependent probe amplification (MLPA) were performed. Bioinformatics analysis was performed with Ingenuity Clinical Insights software (Qiagen). The patient's electronic record was utilized after receiving informed consent. In this report, we present seven cases of HNF1B-associated kidney disease, each featuring distinct genetic abnormalities and displaying diverse extrarenal manifestations. Over 12 years, the mean decline in eGFR averaged -2.22 ± 0.7 mL/min/1.73 m2. Diabetes mellitus was present in five patients, kidney dysplastic lesions in six patients, pancreatic dysplasia, hypomagnesemia and abnormal liver function tests in three patients each. This case series emphasizes the phenotypic variability and the fast decline in kidney function associated with HNF-1B-related disease. Additionally, it underscores that complex clinical presentations may have a retrospectively straightforward explanation through the use of diverse genetic analytical tools.
Collapse
Affiliation(s)
- Ioannis Petrakis
- Department of Nephrology, University of Crete, 71500 Heraklion, Greece; (I.P.); (E.D.); (K.D.); (C.P.); (A.A.)
| | - Maria Sfakiotaki
- Department of Endocrinology, University of Crete, 71500 Heraklion, Greece; (M.S.); (P.X.)
| | - Maria Bitsori
- Department of Pediatrics, University of Crete, 71500 Heraklion, Greece; (M.B.); (E.G.)
| | - Eleni Drosataki
- Department of Nephrology, University of Crete, 71500 Heraklion, Greece; (I.P.); (E.D.); (K.D.); (C.P.); (A.A.)
| | - Kleio Dermitzaki
- Department of Nephrology, University of Crete, 71500 Heraklion, Greece; (I.P.); (E.D.); (K.D.); (C.P.); (A.A.)
| | - Christos Pleros
- Department of Nephrology, University of Crete, 71500 Heraklion, Greece; (I.P.); (E.D.); (K.D.); (C.P.); (A.A.)
| | - Ariadni Androvitsanea
- Department of Nephrology, University of Crete, 71500 Heraklion, Greece; (I.P.); (E.D.); (K.D.); (C.P.); (A.A.)
| | - Dimitrios Samonakis
- Department of Gastroenterology, University of Crete, 71500 Heraklion, Greece;
| | - Amalia Sertedaki
- First Department of Pediatrics, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Paraskevi Xekouki
- Department of Endocrinology, University of Crete, 71500 Heraklion, Greece; (M.S.); (P.X.)
| | - Emmanouil Galanakis
- Department of Pediatrics, University of Crete, 71500 Heraklion, Greece; (M.B.); (E.G.)
| | - Kostas Stylianou
- Department of Nephrology, University of Crete, 71500 Heraklion, Greece; (I.P.); (E.D.); (K.D.); (C.P.); (A.A.)
| |
Collapse
|
17
|
Almutair A, Almulhem B. Semaglutide as a potential therapeutic alternative for HNF1B-MODY: a case study. Front Endocrinol (Lausanne) 2024; 15:1294264. [PMID: 38524636 PMCID: PMC10957750 DOI: 10.3389/fendo.2024.1294264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/31/2024] [Indexed: 03/26/2024] Open
Abstract
Maturity-onset diabetes of the young (MODY) is a grouping of monogenic disorders. It is characterized by dominantly inherited, non-insulin-dependent diabetes. MODY is relatively rare, encompassing up to 3.5% in those diagnosed under 30 years of age. Specific types are most commonly treated with sulfonylurea, particularly those identified as HNF4A-MODY and HNF1A-MODY. HNF1B-MODY is another type that is most frequently managed with insulin therapy but lacks a defined precision treatment. We present an 18-year-old, non-obese female patient diagnosed with HNF1B-MODY. She displays complete gene deletion, a renal cyst, and hypomagnesemia. Her treatment plan includes both long- and short-acting insulin, though she frequently encountered hypoglycemia and hyperglycemia. Semaglutide, a GLP-1RA, was administered weekly over 4 months. The patient's glucose level was continuously tracked using Dexcom's Continuous Glucose Monitoring system. The data suggested a notable improvement in her condition: time-in-range (TIR) increased from 70% to 88%, with some days achieving 100%, and the frequency of hypoglycemic episodes, indicated by time-below-range values, fell from 5% to 1%. The time-above-range values also dropped from 25% to 10%, and her HbA1c levels declined from 7% to 5.6%. During the semaglutide therapy, we were able to discontinue her insulin treatment. Additionally, her body mass index (BMI) was reduced from 24.1 to 20.1 kg/m2. However, the semaglutide treatment was halted after 4 months due to side effects such as nausea, vomiting, and reduced appetite. Other contributing factors included exam stress and a COVID-19 infection, which forced a switch back to insulin. Her last recorded HbA1c level under exclusive insulin therapy rose to 7.1%, and her BMI increased to 24.9 kg/m2. In conclusion, semaglutide could potentially replace insulin to improve glucose variability, TIR, and HbA1c in patients with HNF1B-MODY. However, more extensive studies are required to confirm its long-term safety and efficacy.
Collapse
Affiliation(s)
- Angham Almutair
- Pediatric Department, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdul-Aziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
| | - Beshaier Almulhem
- Pediatric Department, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, King Saud bin Abdul-Aziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia
- College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
18
|
Jankowski J, Lee HK, Liu C, Wilflingseder J, Hennighausen L. Sexually dimorphic renal expression of Klotho is directed by a kidney-specific distal enhancer responsive to HNF1b. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.29.582831. [PMID: 38529500 PMCID: PMC10962737 DOI: 10.1101/2024.02.29.582831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Transcription enhancers are genomic sequences regulating common and tissue-specific genes and their disruption can contribute to human disease development and progression. Klotho, a sexually dimorphic gene specifically expressed in kidney, is well-linked to kidney dysfunction and its deletion from the mouse genome leads to premature aging and death. However, the sexually dimorphic regulation of Klotho is not understood. Here, we characterize two candidate Klotho enhancers using H3K27ac epigenetic marks and transcription factor binding and investigate their functions, individually and combined, through CRISPR-Cas9 genome engineering. We discovered that only the distal (E1), but not the proximal (E2) candidate region constitutes a functional enhancer, with the double deletion not causing Klotho expression to further decrease. E1 activity is dependent on HNF1b transcription factor binding site within the enhancer. Further, E1 controls the sexual dimorphism of Klotho as evidenced by qPCR and RNA-seq. Despite the sharp reduction of Klotho mRNA, unlike germline Klotho knockouts, mutant mice presented normal phenotype, including weight, lifespan, and serum biochemistry. Lastly, only males lacking E1 display more prominent acute, but not chronic kidney injury responses, indicating a remarkable range of potential adaptation to isolated Klotho loss, especially in female E1 knockouts, retaining renoprotection despite over 80% Klotho reduction.
Collapse
Affiliation(s)
- Jakub Jankowski
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Hye Kyung Lee
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Chengyu Liu
- Transgenic Core, National Heart, Lung, and Blood Institute, US National Institutes of Health, Bethesda, MD 20892, USA
| | - Julia Wilflingseder
- Department of Physiology and Pathophysiology, University of Veterinary Medicine Vienna, Veterinärplatz 1, 1210 Vienna, Austria
| | - Lothar Hennighausen
- Section of Genetics and Physiology, Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health, Bethesda, Maryland, 20892, USA
| |
Collapse
|
19
|
Thompson WS, Babayev SN, McGowan ML, Kattah AG, Wick MJ, Bendel-Stenzel EM, Chebib FT, Harris PC, Dahl NK, Torres VE, Hanna C. State of the Science and Ethical Considerations for Preimplantation Genetic Testing for Monogenic Cystic Kidney Diseases and Ciliopathies. J Am Soc Nephrol 2024; 35:235-248. [PMID: 37882743 PMCID: PMC10843344 DOI: 10.1681/asn.0000000000000253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
There is a broad phenotypic spectrum of monogenic polycystic kidney diseases (PKDs). These disorders often involve cilia-related genes and lead to the development of fluid-filled cysts and eventual kidney function decline and failure. Preimplantation genetic testing for monogenic (PGT-M) disorders has moved into the clinical realm. It allows prospective parents to avoid passing on heritable diseases to their children, including monogenic PKD. The PGT-M process involves embryo generation through in vitro fertilization, with subsequent testing of embryos and selective transfer of those that do not harbor the specific disease-causing variant(s). There is a growing body of literature supporting the success of PGT-M for autosomal-dominant and autosomal-recessive PKD, although with important technical limitations in some cases. This technology can be applied to many other types of monogenic PKD and ciliopathies despite the lack of existing reports in the literature. PGT-M for monogenic PKD, like other forms of assisted reproductive technology, raises important ethical questions. When considering PGT-M for kidney diseases, as well as the potential to avoid disease in future generations, there are regulatory and ethical considerations. These include limited government regulation and unstandardized consent processes, potential technical errors, high cost and equity concerns, risks associated with pregnancy for mothers with kidney disease, and the impact on all involved in the process, including the children who were made possible with this technology.
Collapse
Affiliation(s)
- Whitney S. Thompson
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Biomedical Ethics Research Program, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Division of Neonatal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Samir N. Babayev
- Division of Reproductive Endocrinology and Infertility, Mayo Clinic, Rochester, Minnesota
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Michelle L. McGowan
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Biomedical Ethics Research Program, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Andrea G. Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Myra J. Wick
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | | | - Fouad T. Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Neera K. Dahl
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Christian Hanna
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
20
|
Satariano M, Ghose S, Raina R. The Pathophysiology of Inherited Renal Cystic Diseases. Genes (Basel) 2024; 15:91. [PMID: 38254980 PMCID: PMC10815569 DOI: 10.3390/genes15010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Renal cystic diseases (RCDs) can arise from utero to early adulthood and present with a variety of symptoms including renal, hepatic, and cardiovascular manifestations. It is well known that common RCDs such as autosomal polycystic kidney disease and autosomal recessive kidney disease are linked to genes such as PKD1 and PKHD1, respectively. However, it is important to investigate the genetic pathophysiology of how these gene mutations lead to clinical symptoms and include some of the less-studied RCDs, such as autosomal dominant tubulointerstitial kidney disease, multicystic dysplastic kidney, Zellweger syndrome, calyceal diverticula, and more. We plan to take a thorough look into the genetic involvement and clinical sequalae of a number of RCDs with the goal of helping to guide diagnosis, counseling, and treatment.
Collapse
Affiliation(s)
- Matthew Satariano
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (M.S.); (S.G.)
| | - Shaarav Ghose
- Department of Medicine, Northeast Ohio Medical University, Rootstown, OH 44272, USA; (M.S.); (S.G.)
| | - Rupesh Raina
- Akron Nephrology Associates, Cleveland Clinic Akron General Medical Center, Akron, OH 44307, USA
- Department of Nephrology, Akron Children’s Hospital, Akron, OH 44308, USA
| |
Collapse
|
21
|
Hasegawa Y, Takahashi Y, Nagasawa K, Kinno H, Oda T, Hangai M, Odashima Y, Suzuki Y, Shimizu J, Ando T, Egawa I, Hashizume K, Nata K, Yabe D, Horikawa Y, Ishigaki Y. Japanese 17q12 Deletion Syndrome with Complex Clinical Manifestations. Intern Med 2024; 63:687-692. [PMID: 38432894 PMCID: PMC10982014 DOI: 10.2169/internalmedicine.1660-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/29/2023] [Indexed: 03/05/2024] Open
Abstract
17q12 deletion syndrome is a rare chromosomal anomaly with variable phenotypes, caused by the heterozygous deletion of chromosome 17q12. We herein report a 35-year-old Japanese patient with chromosomal 17q12 deletion syndrome identified by de novo deletion of the 1.46 Mb segment at the 17q12 band by genetic analyses. He exhibited a wide range of phenotypes, such as maturity-onset diabetes of the young (MODY) type 5, structural or functional abnormalities of the kidney, liver, and pancreas; facial dysmorphic features, electrolyte disorders; keratoconus, and acquired perforating dermatosis. This case report provides valuable resources concerning the clinical spectrum of rare 17q12 deletion syndrome.
Collapse
Affiliation(s)
- Yutaka Hasegawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Yoshihiko Takahashi
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Kan Nagasawa
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Hirofumi Kinno
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Tomoyasu Oda
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Mari Hangai
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Yoshimi Odashima
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Yoko Suzuki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Jun Shimizu
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Toshihiko Ando
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| | - Isao Egawa
- Department of Ophthalmology, School of Medicine, Iwate Medical University, Japan
| | - Kouhei Hashizume
- Department of Ophthalmology, School of Medicine, Iwate Medical University, Japan
| | - Koji Nata
- Division of Medical Biochemistry, School of Pharmacy, Iwate Medical University, Japan
| | - Daisuke Yabe
- Department of Diabetes, Endocrinology and Metabolism/Department of Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Japan
| | - Yukio Horikawa
- Department of Diabetes, Endocrinology and Metabolism/Department of Rheumatology and Clinical Immunology, Gifu University Graduate School of Medicine, Japan
- Clinical Genetics Center, Gifu University Hospital, Japan
| | - Yasushi Ishigaki
- Division of Diabetes, Metabolism and Endocrinology, Department of Internal Medicine, Iwate Medical University, Japan
| |
Collapse
|
22
|
Vivante A, Tan W, Harrington SG, Udler MS, Pollin TI. Case 36-2023: A 19-Year-Old Man with Diabetes and Kidney Cysts. N Engl J Med 2023; 389:1993-2003. [PMID: 37991859 DOI: 10.1056/nejmcpc2309347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2023]
Affiliation(s)
- Asaf Vivante
- From the Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - both in Israel (A.V.); the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Massachusetts General Hospital, and the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Harvard Medical School - both in Boston; and the Department of Medicine, University of Maryland School of Medicine, Baltimore (T.I.P.)
| | - Weizhen Tan
- From the Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - both in Israel (A.V.); the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Massachusetts General Hospital, and the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Harvard Medical School - both in Boston; and the Department of Medicine, University of Maryland School of Medicine, Baltimore (T.I.P.)
| | - Samantha G Harrington
- From the Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - both in Israel (A.V.); the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Massachusetts General Hospital, and the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Harvard Medical School - both in Boston; and the Department of Medicine, University of Maryland School of Medicine, Baltimore (T.I.P.)
| | - Miriam S Udler
- From the Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - both in Israel (A.V.); the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Massachusetts General Hospital, and the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Harvard Medical School - both in Boston; and the Department of Medicine, University of Maryland School of Medicine, Baltimore (T.I.P.)
| | - Toni I Pollin
- From the Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, and the Faculty of Medicine, Tel Aviv University, Tel Aviv - both in Israel (A.V.); the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Massachusetts General Hospital, and the Departments of Pediatrics (W.T.), Radiology (S.G.H.), and Medicine (M.S.U.), Harvard Medical School - both in Boston; and the Department of Medicine, University of Maryland School of Medicine, Baltimore (T.I.P.)
| |
Collapse
|
23
|
Veerareddy S, Reddy S, Barreto M, Vedherey N, Gopalareddy VV. Increased Liver Enzymes: An Under-Recognized Finding in Maturity-Onset Diabetes of the Young Type 5 (MODY 5). ACG Case Rep J 2023; 10:e01150. [PMID: 37799485 PMCID: PMC10550013 DOI: 10.14309/crj.0000000000001150] [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: 06/11/2023] [Accepted: 08/11/2023] [Indexed: 10/07/2023] Open
Abstract
Maturity-onset diabetes of the young type 5 (MODY 5) is characterized by a single gene mutation in the HNF1B gene. This frequently leads to insulin resistance and presents as young-onset diabetes. Other manifestations can occur in organs expressing hepatocyte nuclear factor-1 beta. This case report highlights family members with MODY 5 presenting with increased liver enzymes with no etiology. The siblings and their mother had a point mutation p.Arg235Trp in HNF1B gene located at 17q12. This variant is associated with autosomal dominant MODY 5 with renal cysts also known as renal cysts and diabetes syndrome.
Collapse
Affiliation(s)
| | - Saigopala Reddy
- University of North Carolina School of Medicine, Chapel Hill, NC
| | | | | | - Vani V. Gopalareddy
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Levine Childrens Hospital at Atrium Health, Charlotte, NC
| |
Collapse
|
24
|
Lauridsen KM, Støy J, Winther-Larsen A, Abildgaard A. Deceived by Elevated A1C: Cases of Misdiagnosed Diabetes. Clin Diabetes 2023; 42:150-155. [PMID: 38230327 PMCID: PMC10788644 DOI: 10.2337/cd23-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Affiliation(s)
- Kasper Munch Lauridsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Julie Støy
- Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Winther-Larsen
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Chemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Abildgaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Chemistry, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
25
|
Hanna C, Iliuta IA, Besse W, Mekahli D, Chebib FT. Cystic Kidney Diseases in Children and Adults: Differences and Gaps in Clinical Management. Semin Nephrol 2023; 43:151434. [PMID: 37996359 DOI: 10.1016/j.semnephrol.2023.151434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Cystic kidney diseases, when broadly defined, have a wide differential diagnosis extending from recessive diseases with a prenatal or pediatric diagnosis, to the most common autosomal-dominant polycystic kidney disease primarily affecting adults, and several other genetic or acquired etiologies that can manifest with kidney cysts. The most likely diagnoses to consider when assessing a patient with cystic kidney disease differ depending on family history, age stratum, radiologic characteristics, and extrarenal features. Accurate identification of the underlying condition is crucial to estimate the prognosis and initiate the appropriate management, identification of extrarenal manifestations, and counseling on recurrence risk in future pregnancies. There are significant differences in the clinical approach to investigating and managing kidney cysts in children compared with adults. Next-generation sequencing has revolutionized the diagnosis of inherited disorders of the kidney, despite limitations in access and challenges in interpreting the data. Disease-modifying treatments are lacking in the majority of kidney cystic diseases. For adults with rapid progressive autosomal-dominant polycystic kidney disease, tolvaptan (V2-receptor antagonist) has been approved to slow the rate of decline in kidney function. In this article, we examine the differences in the differential diagnosis and clinical management of cystic kidney disease in children versus adults, and we highlight the progress in molecular diagnostics and therapeutics, as well as some of the gaps meriting further attention.
Collapse
Affiliation(s)
- Christian Hanna
- Division of Pediatric Nephrology and Hypertension, Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN; Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN.
| | - Ioan-Andrei Iliuta
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Whitney Besse
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Djalila Mekahli
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Jacksonville, FL.
| |
Collapse
|
26
|
Murphy R, Colclough K, Pollin TI, Ikle JM, Svalastoga P, Maloney KA, Saint-Martin C, Molnes J, Misra S, Aukrust I, de Franco A, Flanagan SE, Njølstad PR, Billings LK, Owen KR, Gloyn AL. A Systematic Review of the use of Precision Diagnostics in Monogenic Diabetes. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.15.23288269. [PMID: 37131594 PMCID: PMC10153302 DOI: 10.1101/2023.04.15.23288269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Monogenic forms of diabetes present opportunities for precision medicine as identification of the underlying genetic cause has implications for treatment and prognosis. However, genetic testing remains inconsistent across countries and health providers, often resulting in both missed diagnosis and misclassification of diabetes type. One of the barriers to deploying genetic testing is uncertainty over whom to test as the clinical features for monogenic diabetes overlap with those for both type 1 and type 2 diabetes. In this review, we perform a systematic evaluation of the evidence for the clinical and biochemical criteria used to guide selection of individuals with diabetes for genetic testing and review the evidence for the optimal methods for variant detection in genes involved in monogenic diabetes. In parallel we revisit the current clinical guidelines for genetic testing for monogenic diabetes and provide expert opinion on the interpretation and reporting of genetic tests. We provide a series of recommendations for the field informed by our systematic review, synthesizing evidence, and expert opinion. Finally, we identify major challenges for the field and highlight areas for future research and investment to support wider implementation of precision diagnostics for monogenic diabetes.
Collapse
Affiliation(s)
- Rinki Murphy
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Auckland Diabetes Centre, Te Whatu Ora Health New Zealand, Te Tokai Tumai, Auckland, New Zealand
| | - Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom
| | - Toni I Pollin
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jennifer M Ikle
- Department of Pediatrics, Division of Endocrinology & Diabetes, Stanford School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA
| | - Pernille Svalastoga
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Kristin A Maloney
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Cécile Saint-Martin
- Department of Medical Genetics, AP-HP Pitié-Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Janne Molnes
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Shivani Misra
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Diabetes and Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Ingvild Aukrust
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - aiElisa de Franco
- Department of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Sarah E Flanagan
- Department of Clinical and Biomedical Science, Faculty of Health and Life Sciences, University of Exeter, UK
| | - Pål R Njølstad
- Children and Youth Clinic, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Liana K Billings
- Division of Endocrinology, NorthShore University HealthSystem, Skokie, IL, USA; Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL, USA
| | - Katharine R Owen
- Oxford Center for Diabetes, Endocrinology & Metabolism, University of Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Anna L Gloyn
- Department of Pediatrics, Division of Endocrinology & Diabetes, Stanford School of Medicine, Stanford, CA, USA
- Stanford Diabetes Research Center, Stanford School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford School of Medicine, Stanford, CA, USA
| |
Collapse
|
27
|
De Groof J, Dachy A, Breysem L, Mekahli D. Cystic kidney diseases in children. Arch Pediatr 2023; 30:240-246. [PMID: 37062654 DOI: 10.1016/j.arcped.2023.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/24/2022] [Accepted: 02/12/2023] [Indexed: 04/18/2023]
Abstract
Cystic kidney disease comprises a broad group of heterogeneous diseases, which differ greatly in age at onset, disease manifestation, systemic involvement, disease progression, and long-term prognosis. As our understanding of these diseases continues to evolve and new treatment strategies continue to emerge, correctly differentiating and diagnosing these diseases becomes increasingly important. In this review, we aim to highlight the key features of the most relevant cystic kidney diseases, underscore important diagnostic characteristics of each disease, and present specific management options if applicable.
Collapse
Affiliation(s)
- J De Groof
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - A Dachy
- PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium; Department of Pediatrics, ULiège Academic Hospital, Liège, Belgium
| | - L Breysem
- Department of Pediatric Radiology, University Hospitals Leuven, Leuven, Belgium
| | - D Mekahli
- Department of Pediatric Nephrology, University Hospitals Leuven, Leuven, Belgium; PKD Research Group, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.
| |
Collapse
|
28
|
Nitte CM, Dobelke F, König J, Konrad M, Becker K, Kamp-Becker I, Weber S, for the NEOCYST consortium. Review of neurodevelopmental disorders in patients with HNF1B gene variations. Front Pediatr 2023; 11:1149875. [PMID: 36969268 PMCID: PMC10034397 DOI: 10.3389/fped.2023.1149875] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/20/2023] [Indexed: 03/25/2023] Open
Abstract
This review investigates the association between neurodevelopmental disorders (NDD) and variations of the gene HNF1B. Heterozygous intragenetic mutations or heterozygous gene deletions (17q12 microdeletion syndrome) of HNF1B are the cause of a multi-system developmental disorder, termed renal cysts and diabetes syndrome (RCAD). Several studies suggest that in general, patients with genetic variation of HNF1B have an elevated risk for additional neurodevelopmental disorders, especially autism spectrum disorder (ASD) but a comprehensive assessment is yet missing. This review provides an overview including all available studies of patients with HNF1B mutation or deletion with comorbid NDD with respect to the prevalence of NDDs and in how they differ between patients with an intragenic mutation or 17q12 microdeletion. A total of 31 studies was identified, comprising 695 patients with variations in HNF1B, (17q12 microdeletion N = 416, mutation N = 279). Main results include that NDDs are present in both groups (17q12 microdeletion 25.2% vs. mutation 6.8%, respectively) but that patients with 17q12 microdeletions presented more frequently with any NDDs and especially with learning difficulties compared to patients with a mutation of HNF1B. The observed prevalence of NDDs in patients with HNF1B variations seems to be higher than in the general population, but the validity of the estimated prevalence must be deemed insufficient. This review shows that systematical research of NDDs in patients with HNF1B mutations or deletions is lacking. Further studies regarding neuropsychological characteristics of both groups are needed. NDDs might be a concomitant of HFN1B-related disease and should be considered in clinical routine and scientific reports.
Collapse
Affiliation(s)
- Clara Marie Nitte
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Philipps University, Marburg, Germany
- Correspondence: Clara Nittel
| | - Frederike Dobelke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Philipps University, Marburg, Germany
| | - Jens König
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| | - Martin Konrad
- Department of General Pediatrics, University Children’s Hospital, Münster, Germany
| | - Katja Becker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Philipps University, Marburg, Germany
| | - Inge Kamp-Becker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Philipps University, Marburg, Germany
| | - Stefanie Weber
- Department of Pediatric and Adolescent Medicine, Philipps University, Marburg, Germany
| | | |
Collapse
|
29
|
Vollmer S, Katzman P, Londahl M. HNF1B variant without hyperglycaemia as a cause of isolated profound hypomagnesaemia. BMJ Case Rep 2023; 16:e254274. [PMID: 36759045 PMCID: PMC9923289 DOI: 10.1136/bcr-2022-254274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
A young man presented unconscious with severe hyponatraemia, hypokalaemia, hypomagnesaemia and metabolic alkalosis. After 4 months of treatment in hospital, the hypomagnesaemia persisted. The patient had no signs of diabetes mellitus, and radiology showed no abnormalities of the kidneys, pancreas or genitourinary tract. A parenteral magnesium load demonstrated renal wasting with increased fractional urinary excretion of magnesium. Genetic tests for Gitelman as well as Bartter syndromes were negative. However, a wider genetic panel revealed that the patient was heterozygous for a deletion on chromosome band 17q12, encompassing the whole HNF1B gene.This case highlights the importance of considering pathogenic HNF1B variants in isolated profound hypomagnesaemia caused by renal wasting. Pathogenic HNF1B variants may partly mimic hypomagnesaemia found in Gitelman and Bartter syndromes and may be present without other features linked to HNF1B variants, including diabetes mellitus.
Collapse
Affiliation(s)
- Shobitha Vollmer
- Department of Endocrinology, Skane University Hospital, Lund, Sweden
| | - Per Katzman
- Department of Endocrinology, Skane University Hospital, Lund, Sweden
| | - Magnus Londahl
- Department of Endocrinology, Skane University Hospital, Lund, Sweden
- Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
| |
Collapse
|
30
|
Tholen LE, Latta F, Martens JHA, Hoenderop JGJ, de Baaij JHF. Transcription factor HNF1β controls a transcriptional network regulating kidney cell structure and tight junction integrity. Am J Physiol Renal Physiol 2023; 324:F211-F224. [PMID: 36546837 DOI: 10.1152/ajprenal.00199.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Mutations in the hepatocyte nuclear factor (HNF)1β gene (HNF1B) cause autosomal dominant tubulointerstitial kidney disease, a rare and heterogeneous disease characterized by renal cysts and/or malformation, maturity-onset diabetes of the young, hypomagnesemia, and hypokalemia. The electrolyte disturbances may develop in the distal part of the nephron, which is important for fine-tuning of Mg2+ and Ca2+ reabsorption. Therefore, we aimed to study the transcriptional network directed by HNF1β in the distal part of the nephron. We combined HNF1β chromatin immunoprecipitation-sequencing and mRNA expression data to identify direct targets of HNF1β in a renal distal convoluted tubule cell line (mpkDCT). Gene Ontology term pathway analysis demonstrated enrichment of cell polarity, cell-cell junction, and cytoskeleton pathways in the dataset. Genes directly and indirectly regulated by HNF1β within these pathways included members of the apical and basolateral polarity complexes including Crumbs protein homolog 3 (Crb3), partitioning defective 6 homolog-β (Pard6b), and LLGL Scribble cell polarity complex component 2 (Llgl2). In monolayers of mouse inner medullary collecting duct 3 cells expressing dominant negative Hnf1b, tight junction integrity was compromised, as observed by reduced transepithelial electrical resistance values and increased permeability for fluorescein (0.4 kDa) compared with wild-type cells. Expression of dominant negative Hnf1b also led to a decrease in height (30%) and an increase in surface (58.5%) of cells grown on membranes. Moreover, three-dimensional spheroids formed by cells expressing dominant negative Hnf1b were reduced in size compared with wild-type spheroids (30%). Together, these findings demonstrate that HNF1β directs a transcriptional network regulating tight junction integrity and cell structure in the distal part of the nephron.NEW & NOTEWORTHY Genetic defects in transcription factor hepatocyte nuclear factor (HNF)1β cause a heterogeneous disease characterized by electrolyte disturbances, kidney cysts, and diabetes. By combining RNA-sequencing and HNF1β chromatin immunoprecipitation-sequencing data, we identified new HNF1β targets that were enriched for cell polarity pathways. Newly discovered targets included members of polarity complexes Crb3, Pard6b, and Llgl2. Functional assays in kidney epithelial cells demonstrated decreased tight junction integrity and a loss of typical cuboidal morphology in mutant Hnf1b cells.
Collapse
Affiliation(s)
- Lotte E Tholen
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Femke Latta
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Joost H A Martens
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- 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
| |
Collapse
|
31
|
Gambella A, Kalantari S, Cadamuro M, Quaglia M, Delvecchio M, Fabris L, Pinon M. The Landscape of HNF1B Deficiency: A Syndrome Not Yet Fully Explored. Cells 2023; 12:cells12020307. [PMID: 36672242 PMCID: PMC9856658 DOI: 10.3390/cells12020307] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The hepatocyte nuclear factor 1β (HNF1B) gene is involved in the development of specialized epithelia of several organs during the early and late phases of embryogenesis, performing its function mainly by regulating the cell cycle and apoptosis pathways. The first pathogenic variant of HNF1B (namely, R177X) was reported in 1997 and is associated with the maturity-onset diabetes of the young. Since then, more than 230 different HNF1B variants have been reported, revealing a multifaceted syndrome with complex and heterogenous genetic, pathologic, and clinical profiles, mainly affecting the pediatric population. The pancreas and kidneys are the most frequently affected organs, resulting in diabetes, renal cysts, and a decrease in renal function, leading, in 2001, to the definition of HNF1B deficiency syndrome, including renal cysts and diabetes. However, several other organs and systems have since emerged as being affected by HNF1B defect, while diabetes and renal cysts are not always present. Especially, liver involvement has generally been overlooked but recently emerged as particularly relevant (mostly showing chronically elevated liver enzymes) and with a putative relation with tumor development, thus requiring a more granular analysis. Nowadays, HNF1B-associated disease has been recognized as a clinical entity with a broader and more variable multisystem phenotype, but the reasons for the phenotypic heterogeneity are still poorly understood. In this review, we aimed to describe the multifaceted nature of HNF1B deficiency in the pediatric and adult populations: we analyzed the genetic, phenotypic, and clinical features of this complex and misdiagnosed syndrome, covering the most frequent, unusual, and recently identified traits.
Collapse
Affiliation(s)
- Alessandro Gambella
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Division of Liver and Transplant Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Silvia Kalantari
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | | | - Marco Quaglia
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Maurizio Delvecchio
- Metabolic Disease and Genetics Unit, Giovanni XXIII Children’s Hospital, AOU Policlinico di Bari, 70124 Bari, Italy
- Correspondence:
| | - Luca Fabris
- Department of Molecular Medicine, University of Padova, 35121 Padua, Italy
- Liver Center, Digestive Disease Section, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Michele Pinon
- Pediatric Gastroenterology Unit, Regina Margherita Children’s Hospital, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
32
|
Abstract
Mid-aortic syndrome (MAS) is an uncommon condition characterized by severe narrowing of the abdominal aorta, usually involving visceral and renal arteries. Most patients are asymptomatic and typically present with incidental hypertension which might evolve into end-organ damage if untreated. Our aim was to review 8 new pediatric MAS cases. A retrospective observational study of all pediatric patients with MAS diagnosis (April 1992-November 2021) was conducted. Patients underwent systematic evaluation (medical and family history; 12-lead electrocardiogram; echocardiogram; angiography and/or computed tomography or magnetic resonance angiography). 8 pediatric patients with MAS were included. Median age at diagnosis was 2.6 [0.2-4.7] years; median follow-up time was 8.6 [6.6-10.0] years. 6/8 patients presented with incidental hypertension, 1/8 with heart murmur, and 1/8 with heart failure symptoms. All patients were on antihypertensive treatment. 1/8 patients underwent surgery and 7/8 an endovascular treatment. At the end of the study period, among the 6 patients that underwent a successful endovascular procedure, 2 achieved good blood pressure (BP) control, 2 acceptable BP control, 1 stage 1 hypertension and, another, stage 2 hypertension. There was 1 death during follow-up. BP monitoring in pediatric patients is crucial for early recognition of MAS. Treatment should be based on the individual clinical characteristics of patients with careful planning of surgical revascularisation, if possible, after adult growth is completed. Our study demonstrates that endovascular treatment might be a good alternative to surgery. Nevertheless, further trials with larger sample size and longer-term follow-up are required to determine the best treatment approach.
Collapse
|
33
|
Tholen LE, Schigt H, Kleuskens SGE, Bos C, Spruijt CG, Willemsen B, Vermeulen M, Hoenderop JGJ, de Baaij JHF. HNF1β-associated cyst development and electrolyte disturbances are not explained by BAIAP2L2 expression. FASEB J 2023; 37:e22696. [PMID: 36520027 DOI: 10.1096/fj.202201121r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/21/2022] [Accepted: 11/28/2022] [Indexed: 12/16/2022]
Abstract
Mutations or deletions in transcription factor hepatocyte nuclear factor 1 homeobox β (HNF1β) cause renal cysts and/or malformation, maturity-onset diabetes of the young and electrolyte disturbances. Here, we applied a comprehensive bioinformatic approach on ChIP-seq, RNA-seq, and gene expression array studies to identify novel transcriptional targets of HNF1β explaining the kidney phenotype of HNF1β patients. We identified BAR/IMD Domain Containing Adaptor Protein 2 Like 2 (BAIAP2L2), as a novel transcriptional target of HNF1β and validated direct transcriptional activation of the BAIAP2L2 promoter by a reporter luciferase assay. Using mass spectrometry analysis, we show that BAIAP2L2 binds to other members of the I-BAR domain-containing family: BAIAP2 and BAIAP2L1. Subsequently, the role of BAIAP2L2 in maintaining epithelial cell integrity in the kidney was assessed using Baiap2l2 knockout cell and mouse models. Kidney epithelial cells lacking functional BAIAP2L2 displayed normal F-actin distribution at cell-cell contacts and formed polarized three-dimensional spheroids with a lumen. In vivo, Baiap2l2 knockout mice displayed normal kidney and colon tissue morphology and serum and urine electrolyte concentrations were not affected. Altogether, our study is the first to characterize the function of BAIAP2L2 in the kidney in vivo and we report that mice lacking BAIAP2L2 exhibit normal electrolyte homeostasis and tissue morphology under physiological conditions.
Collapse
Affiliation(s)
- Lotte E Tholen
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Heidi Schigt
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sanne G E Kleuskens
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Caro Bos
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Cornelia G Spruijt
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Brigith Willemsen
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen, The Netherlands
| | - Joost G J Hoenderop
- 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
| |
Collapse
|
34
|
Molina LM, Salgado CM, Reyes-Múgica M. Potter Deformation Sequence Caused by 17q12 Deletion: A Lethal Constellation. Pediatr Dev Pathol 2022; 26:144-148. [PMID: 36513606 DOI: 10.1177/10935266221139341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
17q12 deletion syndrome causes developmental abnormalities of the kidneys, pancreas, genital tract, and neurodevelopment, and it has a wide range of phenotypes ranging from fetal demise to normal adulthood with minimal renal impairment. Here we describe a rare case of 17q12 deletion diagnosed prenatally, complicated by anhydramnios and Potter sequence. The baby was born but necessitated life-saving interventions due to pulmonary and renal insufficiency and ultimately succumbed to multi-organ failure. We present full autopsy results describing findings linked to 17q12 deletion, including severe bilateral multicystic renal dysplasia, pancreatic hypoplasia, and cysts adjacent to the Fallopian tubes. We also describe pulmonary hypoplasia and Potter facies as consequences of anhydramnios. We correlate these findings to our current understanding of molecular signals altered by 17q12 deletion, notably affecting HNF1B and LHX1 genes, which are known to mediate renal and genitourinary tract development.
Collapse
Affiliation(s)
- Laura M Molina
- Department of Pathology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia M Salgado
- Department of Pathology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Miguel Reyes-Múgica
- Department of Pathology, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
35
|
Cao B, Liu M, Zhang Y, Chen J, Li X, Su C, Yang W, Liu M, Wu D, Li W, Liang X, Wang Q, Wei H, Gong C. An effective preselection criterion for MODY with an increasingly positive genetic testing rate by NGS: results from two cohorts of Chinese children. Am J Physiol Endocrinol Metab 2022; 323:E529-E534. [PMID: 36383636 DOI: 10.1152/ajpendo.00171.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The purpose of this study was to determine the frequency of maturity-onset diabetes of the young (MODY) in two selected cohorts of Chinese children with diabetes and clinically suspected MODY, using next-generation sequencing (NGS). Ninety-three children who met the comprehensive criteria of suspected MODY were enrolled in two cohorts. A custom NGS panel or a whole exon group was used for sequencing. We identified 55/93 (59.1%) children with pathogenic and likely pathogenic MODY variants. Forty-two (76.3%) were confirmed to have the GCK (MODY2) mutation. Additionally, five had the HNF1A (MODY3), two the HNF1B (MODY5), one the 17q12 microdeletion (MODY5), two the HNF4A (MODY1), two the ABCC8 (MODY12), and one the PDX1 mutation (MODY4). Of these, 13 novel variants were detected in different genes. By comparing the gene-positive with gene-negative children, we found that discriminatory factors for MODY at diagnosis included lower HbA1c [7.4% vs. 10.2% (53 vs. 86 mmol/mol); P = 0.002], lower body mass index z score (0.2 vs. 1.0; P = 0.01), lower onset age (8.1 vs. 11.2 years; P = 0.001), and lower C-peptide (1.4 vs. 2.5 ng/mL; P = 0.02). In conclusion, the criteria used in this study for screening MODY are effective, and MODY2 is the most common subtype (76%), followed by MODY3 and MODY5. Some rare MODY subtypes have been reported in Chinese children.NEW & NOTEWORTHY We proved the clinical suspicion of maturity-onset diabetes of the young (MODY) according to the comprehensive criterion for next-generation sequencing testing, which helps to identify both common and rare MODYs, leading to accurate diagnosis and personalized treatment.
Collapse
Affiliation(s)
- Bingyan Cao
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Meijuan Liu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yingxian Zhang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Jiajia Chen
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xiaoqiao Li
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Chang Su
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Yang
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Min Liu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Di Wu
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenjing Li
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xuejun Liang
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qiao Wang
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Haiyan Wei
- Department of Endocrinology, Genetics and Metabolism, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, China
| | - Chunxiu Gong
- Department of Endocrinology, Genetics and Metabolism, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
36
|
Kagan M, Pleniceanu O, Vivante A. The genetic basis of congenital anomalies of the kidney and urinary tract. Pediatr Nephrol 2022; 37:2231-2243. [PMID: 35122119 DOI: 10.1007/s00467-021-05420-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 10/19/2022]
Abstract
During the past decades, remarkable progress has been made in our understanding of the molecular basis of kidney diseases, as well as in the ability to pinpoint disease-causing genetic changes. Congenital anomalies of the kidney and urinary tract (CAKUT) are remarkably diverse, and may be either isolated to the kidney or involve other systems, and are notorious in their variable genotype-phenotype correlations. Genetic conditions underlying CAKUT are individually rare, but collectively contribute to disease etiology in ~ 16% of children with CAKUT. In this review, we will discuss basic concepts of kidney development and genetics, common causes of monogenic CAKUT, and the approach to diagnosing and managing a patient with suspected monogenic CAKUT. Altogether, the concepts presented herein represent an introduction to the emergence of nephrogenetics, a fast-growing multi-disciplinary field that is focused on deciphering the causes and manifestations of genetic kidney diseases as well as providing the framework for managing patients with genetic forms of CAKUT.
Collapse
Affiliation(s)
- Maayan Kagan
- Pediatric Department B and Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Kidney Research Lab, The Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Asaf Vivante
- Pediatric Department B and Pediatric Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601, Ramat Gan, Israel. .,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel. .,Talpiot Medical Leadership Program, Tel HaShomer, Ramat Gan, Israel.
| |
Collapse
|
37
|
Živná M, Kidd KO, Barešová V, Hůlková H, Kmoch S, Bleyer AJ. Autosomal dominant tubulointerstitial kidney disease: A review. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:309-324. [PMID: 36250282 PMCID: PMC9619361 DOI: 10.1002/ajmg.c.32008] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 09/10/2022] [Accepted: 09/29/2022] [Indexed: 01/11/2023]
Abstract
The clinical characteristics of autosomal dominant tubulointerstitial kidney disease (ADTKD) include bland urinary sediment, slowly progressive chronic kidney disease (CKD) with many patients reaching end stage renal disease (ESRD) between age 20 and 70 years, and autosomal dominant inheritance. Due to advances in genetic diagnosis, ADTKD is becoming increasingly recognized as a cause of CKD. Pathogenic variants in UMOD, MUC1, and REN are the most common causes of ADTKD. ADTKD-UMOD is also associated with hyperuricemia and gout. ADTKD-REN often presents in childhood with mild hypotension, CKD, hyperkalemia, acidosis, and anemia. ADTKD-MUC1 patients present only with CKD. This review describes the pathophysiology, genetics, clinical manifestation, and diagnosis for ADTKD, with an emphasis on genetic testing and genetic counseling suggestions for patients.
Collapse
Affiliation(s)
- Martina Živná
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Kendrah O. Kidd
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Wake Forest University School of MedicineSection on NephrologyWinston‐SalemNorth CarolinaUSA
| | - Veronika Barešová
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Helena Hůlková
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Stanislav Kmoch
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Wake Forest University School of MedicineSection on NephrologyWinston‐SalemNorth CarolinaUSA
| | - Anthony J. Bleyer
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of MedicineCharles UniversityPragueCzech Republic
- Wake Forest University School of MedicineSection on NephrologyWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
38
|
Stein D, McNamara E. Congenital Anomalies of the Kidneys and Urinary Tract. Clin Perinatol 2022; 49:791-798. [PMID: 36113935 DOI: 10.1016/j.clp.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Congenital anomalies of the kidneys and urinary tract (CAKUT) are some of the most common abnormalities detected on prenatal imaging assessment. It is estimated that CAKUT comprises 20% to 30% of all major birth defects. More than 200 clinical syndromes currently include CAKUT as a component of the phenotype. This chapter outlines the evaluation and management of the most common forms of CAKUT.
Collapse
Affiliation(s)
- Deborah Stein
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Erin McNamara
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Tholen LE, Hoenderop JGJ, de Baaij JHF. Mechanisms of ion transport regulation by HNF1β in the kidney: beyond transcriptional regulation of channels and transporters. Pflugers Arch 2022; 474:901-916. [PMID: 35554666 PMCID: PMC9338905 DOI: 10.1007/s00424-022-02697-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 01/01/2023]
Abstract
Hepatocyte nuclear factor 1β (HNF1β) is a transcription factor essential for the development and function of the kidney. Mutations in and deletions of HNF1β cause autosomal dominant tubule interstitial kidney disease (ADTKD) subtype HNF1β, which is characterized by renal cysts, diabetes, genital tract malformations, and neurodevelopmental disorders. Electrolyte disturbances including hypomagnesemia, hyperuricemia, and hypocalciuria are common in patients with ADTKD-HNF1β. Traditionally, these electrolyte disturbances have been attributed to HNF1β-mediated transcriptional regulation of gene networks involved in ion transport in the distal part of the nephron including FXYD2, CASR, KCNJ16, and FXR. In this review, we propose additional mechanisms that may contribute to the electrolyte disturbances observed in ADTKD-HNF1β patients. Firstly, kidney development is severely affected in Hnf1b-deficient mice. HNF1β is required for nephron segmentation, and the absence of the transcription factor results in rudimentary nephrons lacking mature proximal tubule, loop of Henle, and distal convoluted tubule cluster. In addition, HNF1β is proposed to be important for apical-basolateral polarity and tight junction integrity in the kidney. Interestingly, cilia formation is unaffected by Hnf1b defects in several models, despite the HNF1β-mediated transcriptional regulation of many ciliary genes. To what extent impaired nephron segmentation, apical-basolateral polarity, and cilia function contribute to electrolyte disturbances in HNF1β patients remains elusive. Systematic phenotyping of Hnf1b mouse models and the development of patient-specific kidney organoid models will be essential to advance future HNF1β research.
Collapse
Affiliation(s)
- Lotte E Tholen
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Jeroen H F de Baaij
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, P. O. Box 9101, Nijmegen, 6500 HB, The Netherlands.
| |
Collapse
|
40
|
Bleyer AJ, Wolf MT, Kidd KO, Zivna M, Kmoch S. Autosomal dominant tubulointerstitial kidney disease: more than just HNF1β. Pediatr Nephrol 2022; 37:933-946. [PMID: 34021396 PMCID: PMC8722360 DOI: 10.1007/s00467-021-05118-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/14/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022]
Abstract
Autosomal dominant tubulointerstitial kidney disease (ADTKD) refers to a group of disorders with a bland urinary sediment, slowly progressive chronic kidney disease (CKD), and autosomal dominant inheritance. Due to advances in genetic diagnosis, ADTKD is becoming increasingly recognized as a cause of CKD in both children and adults. ADTKD-REN presents in childhood with mild hypotension, CKD, hyperkalemia, acidosis, and anemia. ADTKD-UMOD is associated with gout and CKD that may present in adolescence and slowly progresses to kidney failure. HNF1β mutations often present in childhood with anatomic abnormalities such as multicystic or dysplastic kidneys, as well as CKD and a number of other extra-kidney manifestations. ADTKD-MUC1 is less common in childhood, and progressive CKD is its sole clinical manifestation, usually beginning in the late teenage years. This review describes the pathophysiology, genetics, clinical characteristics, diagnosis, and treatment of the different forms of ADTKD, with an emphasis on diagnosis. We also present data on kidney function in children with ADTKD from the Wake Forest Rare Inherited Kidney Disease Registry.
Collapse
Affiliation(s)
- Anthony J Bleyer
- Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA.
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Matthias T Wolf
- Pediatric Nephrology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390-0936, USA
| | - Kendrah O Kidd
- Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Martina Zivna
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Stanislav Kmoch
- Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, 27157, USA
- Research Unit of Rare Diseases, Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
41
|
Cui T, Ju HB, Liu PF, Ma YJ, Zhang FX. A case report of CAT gene and HNF1β gene variations in a patient with early-onset diabetes. Open Life Sci 2022; 17:344-350. [PMID: 35480487 PMCID: PMC8989158 DOI: 10.1515/biol-2022-0026] [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: 04/26/2021] [Revised: 11/16/2021] [Accepted: 01/03/2022] [Indexed: 11/15/2022] Open
Abstract
Abstract
Complex forms of diabetes are the ultimate common pathway involving multiple genetic variations and multiple environmental factors. Type 2 diabetes (T2DM) is classified as complex diabetes. Varying degrees of insulin deficiency and tissue insulin resistance are two key links to T2DM. The islet β cell dysfunction plays a crucial role in the pathogenesis of T2DM. The decompensation of the islet β cell to insulin resistance is a common mechanism leading to the pathogenesis of T2DM. Available data show that genetic factors mainly affect cell function. At present, a number of susceptibility genes related to T2DM have been reported at home and abroad. In this study, the diabetes-related genes in the case of early-onset diabetes with a significant family history were examined, and our results showed the presence of the intron mutations of catalase (CAT) gene and hepatocyte nuclear factor 1β (HNF1β) gene. The patient enrolled in this study was observed and analyzed, thus, increasing further understanding of the genes associated with diabetes and exploring the pathogenesis of diabetes from the molecular level. This is significant for guiding the prevention, treatment, and prognosis evaluation of diabetes.
Collapse
Affiliation(s)
- Tao Cui
- Department of Endocrine, 920th Hospital of Joint Logistics Support Force of the Chinese People’s Libration Army , Xishan District , Kunming 650032 , China
| | - Hai-Bing Ju
- Department of Endocrine, 920th Hospital of Joint Logistics Support Force of the Chinese People’s Libration Army , Xishan District , Kunming 650032 , China
| | - Peng-Fei Liu
- Department of Ophthalmology, Western Theater Command Air Force Hospital , Chengdu 610000 , China
| | - Yun-Jun Ma
- Department of Psychiatry, 920th Hospital of Joint Logistics Support Force of the Chinese People’s Libration Army , Kunming 650032 , China
| | - Fu-Xian Zhang
- Department of Endocrine, 920th Hospital of Joint Logistics Support Force of the Chinese People’s Libration Army , Xishan District , Kunming 650032 , China
| |
Collapse
|
42
|
Cujba AM, Alvarez-Fallas ME, Pedraza-Arevalo S, Laddach A, Shepherd MH, Hattersley AT, Watt FM, Sancho R. An HNF1α truncation associated with maturity-onset diabetes of the young impairs pancreatic progenitor differentiation by antagonizing HNF1β function. Cell Rep 2022; 38:110425. [PMID: 35235779 PMCID: PMC8905088 DOI: 10.1016/j.celrep.2022.110425] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 09/23/2021] [Accepted: 02/02/2022] [Indexed: 01/16/2023] Open
Abstract
The HNF1αp291fsinsC truncation is the most common mutation associated with maturity-onset diabetes of the young 3 (MODY3). Although shown to impair HNF1α signaling, the mechanism by which HNF1αp291fsinsC causes MODY3 is not fully understood. Here we use MODY3 patient and CRISPR/Cas9-engineered human induced pluripotent stem cells (hiPSCs) grown as 3D organoids to investigate how HNF1αp291fsinsC affects hiPSC differentiation during pancreatic development. HNF1αp291fsinsC hiPSCs shows reduced pancreatic progenitor and β cell differentiation. Mechanistically, HNF1αp291fsinsC interacts with HNF1β and inhibits its function, and disrupting this interaction partially rescues HNF1β-dependent transcription. HNF1β overexpression in the HNF1αp291fsinsC patient organoid line increases PDX1+ progenitors, while HNF1β overexpression in the HNF1αp291fsinsC patient iPSC line partially rescues β cell differentiation. Our study highlights the capability of pancreas progenitor-derived organoids to model disease in vitro. Additionally, it uncovers an HNF1β-mediated mechanism linked to HNF1α truncation that affects progenitor differentiation and could explain the clinical heterogeneity observed in MODY3 patients. MODY3 patient and CRISPR/Cas9 HNF1αp291fsinsC mutated iPSC lines are generated Mutant iPSCs show deficient pancreatic progenitor and β cell differentiation Mutant truncated HNF1α protein binds wild-type HNF1β protein to hinder its function HNF1β overexpression in MODY3 iPSC line partially rescues β cell differentiation
Collapse
Affiliation(s)
- Ana-Maria Cujba
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | | | | | | | | | | | - Fiona M Watt
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK
| | - Rocio Sancho
- Centre for Stem Cells and Regenerative Medicine, King's College London, London, UK; Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
43
|
Heidenreich LS, Bendel-Stenzel EM, Harris PC, Hanna C. Genetic Etiologies, Diagnosis, and Management of Neonatal Cystic Kidney Disease. Neoreviews 2022; 23:e175-e188. [PMID: 35229136 DOI: 10.1542/neo.23-3-e175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Fetal kidney development is a complex and carefully orchestrated process. The proper formation of kidney tissue involves many transcription factors and signaling pathways. Pathogenic variants in the genes that encodethese factors and proteins can result in neonatal cystic kidney disease. Advancements in genomic sequencing have allowed us to identify many of these variants and better understand the genetic underpinnings for an increasing number of presentations of childhood kidney disorders. This review discusses the genes essential in kidney development, particularly those involved in the structure and function of primary cilia, and implications of gene identification for prognostication and management of cystic kidney disorders.
Collapse
Affiliation(s)
- Leah S Heidenreich
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN
| | - Ellen M Bendel-Stenzel
- Division of Neonatology, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, MN
| | - Peter C Harris
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Christian Hanna
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN
- Division of Pediatric Nephrology and Hypertension, Department of Pediatrics and Adolescent Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
44
|
Le Collen L, Delemer B, Spodenkiewicz M, Cornillet Lefebvre P, Durand E, Vaillant E, Badreddine A, Derhourhi M, Mouhoub TA, Jouret G, Juttet P, Souchon PF, Vaxillaire M, Froguel P, Bonnefond A, Doco Fenzy M. Compound genetic etiology in a patient with a syndrome including diabetes, intellectual deficiency and distichiasis. Orphanet J Rare Dis 2022; 17:86. [PMID: 35227307 PMCID: PMC8887189 DOI: 10.1186/s13023-022-02248-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 02/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We studied a young woman with atypical diabetes associated with mild intellectual disability, lymphedema distichiasis syndrome (LDS) and polymalformative syndrome including distichiasis. We used different genetic tools to identify causative pathogenic mutations and/or copy number variations. RESULTS Although proband's, diabetes mellitus occurred during childhood, type 1 diabetes was unlikely due to the absence of detectable autoimmunity. DNA microarray analysis first identified a de novo, heterozygous deletion at the chr16q24.2 locus. Previously, thirty-three pathogenic or likely pathogenic deletions encompassing this locus have been reported in patients presenting with intellectual deficiency, obesity and/or lymphedema but not with diabetes. Of note, the deletion encompassed two topological association domains, whose one included FOXC2 that is known to be linked with LDS. Via whole-exome sequencing, we found a heterozygous, likely pathogenic variant in WFS1 (encoding wolframin endoplasmic reticulum [ER] transmembrane glycoprotein) which was inherited from her father who also had diabetes. WFS1 is known to be involved in monogenic diabetes. We also found a likely pathogenic variant in USP9X (encoding ubiquitin specific peptidase 9 X-linked) that is involved in X-linked intellectual disability, which was inherited from her mother who had dyscalculia and dyspraxia. CONCLUSIONS Our comprehensive genetic analysis suggested that the peculiar phenotypes of our patient were possibly due to the combination of multiple genetic causes including chr16q24.2 deletion, and two likely pathogenic variants in WFS1 and USP9X.
Collapse
Affiliation(s)
- Lauriane Le Collen
- Department of Endocrinology Diabetology, University Hospital Center of Reims, Reims, France. .,Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France. .,University of Lille, Lille, France. .,Department of Genetic, University Hospital Center of Reims, Reims, France.
| | - Brigitte Delemer
- Department of Endocrinology Diabetology, University Hospital Center of Reims, Reims, France. .,Faculty of Medicine of Reims, CRESTIC EA 3804, University of Reims Champagne Ardenne, Moulin de La Housse, BP 1039, 51687, Reims Cedex 2, France.
| | | | | | - Emmanuelle Durand
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France.,University of Lille, Lille, France
| | - Emmanuel Vaillant
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France.,University of Lille, Lille, France
| | - Alaa Badreddine
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France.,University of Lille, Lille, France
| | - Mehdi Derhourhi
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France.,University of Lille, Lille, France
| | - Tarik Ait Mouhoub
- Department of Genetic, University Hospital Center of Reims, Reims, France
| | - Guillaume Jouret
- Department of Genetic, University Hospital Center of Reims, Reims, France.,Departement of Genetic, 1 rue Louis Rech Dudelange, 3555, Luxembourg, Luxembourg
| | | | | | - Martine Vaxillaire
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France.,University of Lille, Lille, France
| | - Philippe Froguel
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France. .,University of Lille, Lille, France.
| | - Amélie Bonnefond
- Inserm/CNRS UMR 1283/8199, Pasteur Institute of Lille, EGID, Lille, France. .,University of Lille, Lille, France.
| | - Martine Doco Fenzy
- Department of Genetic, University Hospital Center of Reims, Reims, France. .,Faculty of Medicine of Reims, EA 3801, URCA, Reims, France.
| |
Collapse
|
45
|
Patil PA, Taddei T, Jain D, Zhang X. HNF-1β is a More Sensitive and Specific Marker Than C-Reactive Protein for Identifying Biliary Differentiation in Primary Hepatic Carcinomas. Arch Pathol Lab Med 2022; 146:220-226. [PMID: 34086854 DOI: 10.5858/arpa.2020-0725-oa] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2021] [Indexed: 02/05/2023]
Abstract
CONTEXT.— Intrahepatic cholangiocarcinoma (iCCA) needs to be distinguished from hepatocellular carcinoma (HCC) and metastasis, and in the absence of any specific biliary markers, is often a diagnosis of exclusion. Hepatocyte nuclear factor (HNF)-1β is a transcription factor that plays a critical role in bile duct system morphogenesis. OBJECTIVE.— To investigate the diagnostic value of HNF-1β to differentiate iCCA from HCC by immunohistochemistry and compare HNF-1β with C-reactive protein (CRP), a previously identified marker for iCCA. DESIGN.— Cases of iCCA (n = 75), combined hepatocellular-cholangiocarcinoma (cHCC-CCA) (n = 13) and HCC (n = 65) were included in the study. RESULTS.— All cases of iCCA (74 of 74, 100%) expressed HNF-1β compared with CRP expressed in 72.60% (53 of 73). The sensitivity and specificity of HNF-1β to differentiate iCCA from HCC was 100% and 92.31%, whereas the sensitivity and specificity for CRP was 75.58% and 7.79%. The expression of HNF-1β was greater in iCCA and the CCA component of cHCC-CCA compared with CRP (87 of 87, 100% versus 65 of 86, 75.58%; P < .001). On the contrary, CRP was more frequently expressed compared with HNF-1β in HCC and HCC component of cHCC-CCA (71 of 77, 92.21% versus 6 of 78, 7.69%; P < .001). CONCLUSIONS.— Our data indicate that HNF-1β is a more sensitive and specific marker than CRP for the diagnosis of iCCA and to identify the CCA component in cHCC-CCA. Lack of HNF-1β expression may be used to exclude iCCA from consideration in cases of adenocarcinomas of unknown primary.
Collapse
Affiliation(s)
- Pallavi A Patil
- From the Department of Pathology (Patil, Jain, Zhang)
- Patil is currently located in the Department of Pathology at the University of South Alabama, Mobile, Alabama
| | - Tamar Taddei
- Section of Digestive Diseases (Taddei), Yale University School of Medicine, New Haven, Connecticut
| | - Dhanpat Jain
- From the Department of Pathology (Patil, Jain, Zhang)
| | - Xuchen Zhang
- From the Department of Pathology (Patil, Jain, Zhang)
| |
Collapse
|
46
|
Hay E, Cullup T, Barnicoat A. A practical approach to the genomics of kidney disorders. Pediatr Nephrol 2022; 37:21-35. [PMID: 33675412 DOI: 10.1007/s00467-021-04995-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/30/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023]
Abstract
Rapid technological advances in genomic testing continue to increase our understanding of the genetic basis of a wide range of kidney disorders. Establishing a molecular diagnosis benefits the individual by bringing an end to what is often a protracted diagnostic odyssey, facilitates accurate reproductive counselling for families and, in the future, is likely to lead to the delivery of more targeted management and surveillance regimens. The selection of the most appropriate testing modality requires an understanding both of the technologies available and of the genetic architecture and heterogeneity of kidney disease. Whilst we are witnessing a far greater diagnostic yield with broader genetic testing, such approaches invariably generate variants of uncertain significance and secondary incidental findings, which are not only difficult to interpret but present ethical challenges with reporting and feeding back to patients and their families. Here, we review the spectrum of nephrogenetic disorders, consider the optimal approach to genetic testing, explore the clinical utility of obtaining a molecular diagnosis, reflect on the challenges of variant interpretation and look to the future of this dynamic field.
Collapse
Affiliation(s)
- Eleanor Hay
- Department of Clinical Genetics, Great Ormond Street Hospital, London, UK.
| | - Thomas Cullup
- North Thames Genomic Laboratory Hub, Great Ormond Street Hospital, London, UK
| | - Angela Barnicoat
- Department of Clinical Genetics, Great Ormond Street Hospital, London, UK
| |
Collapse
|
47
|
Ge S, Yang M, Cui Y, Wu J, Xu L, Dong J, Liao L. The Clinical Characteristics and Gene Mutations of Maturity-Onset Diabetes of the Young Type 5 in Sixty-One Patients. Front Endocrinol (Lausanne) 2022; 13:911526. [PMID: 35846334 PMCID: PMC9281895 DOI: 10.3389/fendo.2022.911526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022] Open
Abstract
AIMS Maturity-onset diabetes of the young type 5 (MODY5), a rare disease, is very easy to be misdiagnosed as type 2 diabetes. To get better understanding of the disease, we analyzed the clinical characteristics and gene mutations of MODY5. METHODS PubMed, Cochrane, the China National Knowledge Infrastructure, and Wanfang were searched with the following search terms: "MODY5" OR "HNF1B maturity-onset diabetes of the young" OR "maturity-onset diabetes of the young type 5" OR "renal cysts and diabetes syndrome". Clinical characteristics and gene mutations of MODY5 were analyzed. The demography, clinical characteristics, and blood indicators of patients were described utilizing simple summary statistics. Variables were analyzed by t-test, Wilcoxon signed rank test, and Fisher exact test. Spearman's correlation analysis was used for bi-variate analysis. All tests were two-sided, and a p-value < 0.05 was considered statistically significant. Statistical analysis was performed using the Statistical Package for the Social Sciences version 26 for Windows (SPSS). RESULTS A total of 48 literatures were included in this study, including 61 eligible patients and 4 different mutations. Of the 39 patients with available body weight index, 15 (38.46%) were underweight, 21 (53.85%) were normal weight and 3 (7.69%) were overweight or obese. Of the 38 patients with available family history, 25 (65.79%) reported a family history of diabetes. Of the 34 patients with available age of diabetes diagnosis, the median age of diabetes diagnosis was 16.00 years old and 88.24% (30/34) of patients were under 25 years old when they were first diagnosed with diabetes. Renal cysts were presented in 72.41%, hypomagnesemia in 91.67%, and pancreatic dysplasia in 71.88% of the patients. Patients with hepatocyte nuclear factor 1B (HNF1B) deletion had lower serum magnesium, serum creatinine, and higher eGFR than patients with other gene mutations, and the difference was statistically significant. CONCLUSIONS The young onset of diabetes with low or normal BMI, renal cysts, hypomagnesemia, and pancreatic dysplasia should be recommended to genetic testing in order to differentiate MODY5 from other types of diabetes earlier.
Collapse
Affiliation(s)
- Shenghui Ge
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Mengge Yang
- Cheeloo College of Medicine, Shandong University, Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, China
| | - Yuying Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jing Wu
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Lusi Xu
- Cheeloo College of Medicine, Shandong University, Department of Endocrinology and Metabology, Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, China
| | - Jianjun Dong
- Division of Endocrinology, Department of Internal Medicine, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Lin Liao, ; Jianjun Dong,
| | - Lin Liao
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
- Department of Endocrinology and Metabology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational medicine, Shandong Institute of Nephrology, Jinan, China
- *Correspondence: Lin Liao, ; Jianjun Dong,
| |
Collapse
|
48
|
Goknar N, Ekici Avci M, Uckardes D, Kelesoglu E, Tekkus Ermis K, Candan C. Hepatocyte Nuclear Factor 1 Beta Mutation-associated Newborn Onset of Glomerulocystic Kidney Disease: A Case Presentation. Medeni Med J 2021; 36:352-355. [PMID: 34939403 PMCID: PMC8694168 DOI: 10.4274/mmj.galenos.2021.02686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Mutations in hepatocyte nuclear factor-1 beta (HNF1B) are the most commonly identified genetic cause of renal malformations. Heterozygous mutations are associated with renal cysts and diabetes syndrome. Various renal developmental abnormalities and maturity-onset diabetes of the young could be the presenting factors of these mutations. A 10-year-old boy who was evaluated for bilateral cystic kidneys and chronic kidney disease from the newborn period was diagnosed with HNF1B-related glomerulocystic disease by DNA sequencing. The differential diagnosis of autosomal dominant polycystic kidney disease was a diagnostic pitfall. The genetic screening of the family revealed his mother, sister, and brother to have the same mutation. Therefore, genetic diagnosis and counseling are important for cystic kidney diseases not only for formulating the diagnosis and early management plan but also for the diagnosis of potential asymptomatic cases in the family.
Collapse
Affiliation(s)
- Nilufer Goknar
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatric Nephrology, Istanbul, Turkey
| | - Melda Ekici Avci
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatrics, Istanbul, Turkey
| | - Diana Uckardes
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatric Nephrology, Istanbul, Turkey
| | - Emre Kelesoglu
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatric Nephrology, Istanbul, Turkey
| | - Kubra Tekkus Ermis
- University of Health Sciences Turkey, Umraniye Training and Research Hospital, Clinic of Genetics, Istanbul, Turkey
| | - Cengiz Candan
- Istanbul Medeniyet University Faculty of Medicine, Department of Pediatric Nephrology, Istanbul, Turkey
| |
Collapse
|
49
|
Cleper R, Reches A, Shapira D, Simchoni S, Reisman L, Ben-Sira L, Yaron Y, Wolman I, Malinger G, Brabbing-Goldstein D, Ben-Shachar S. Improving renal phenotype and evolving extra-renal features of 17q12 deletion encompassing the HNF1B gene. Transl Pediatr 2021; 10:3130-3139. [PMID: 35070826 PMCID: PMC8753471 DOI: 10.21037/tp-21-386] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/01/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND HNF1B deletion/intragenic mutations are the most commonly identified genetic cause of congenital anomalies of the kidney and urinary tract (CAKUT) suggested by fetal ultrasound findings such as: parenchymal hyperechogenicity, overt cystic changes or gross morphological urinary system (UT) abnormalities. The postnatal evolution of these 17q12 deletions encompassing the HNF1B gene-associated findings has not been assessed in depth. METHODS In this observational study, we present postnatal follow-up findings in 5 of 6 cases (one pregnancy was terminated on parental request) of fetal-onset cystic/hyperechogenic kidneys eventually diagnosed with 17q12 microdeletion encompassing the HNF1B gene between 2009 and 2017. RESULTS Complete normalization of kidney parenchymal abnormalities and of depressed neonatal renal function was observed in 4/5 and 5/5 patients within 2-4.9 years and 1.5-8 months, respectively. All 5 patients had preserved normal renal function at 3-11 years of follow-up. The evolving later-onset renal features included: hypomagnesemia, hyperuricemia, urinary tract infection (UTI), and bilateral grade 3-4 vesicoureteral reflux and bladder diverticula in 3, 3, 2, and 1 patient, respectively. HNF1B gene deletion-associated extra-renal manifestations with delayed presentation were global developmental delay/autistic spectrum disorder (ASD), rolandic-type seizures, overweight, and borderline fasting hyperglycemia observed in 1-2 patients each. Family history was positive for small-size or asymptomatic cystic kidneys with normal function, diabetes mellitus, seizures, and mental/psychiatric problems in 3/6 cases. CONCLUSIONS Fetal-onset HNF1B deletion-associated kidneys' parenchymal abnormalities confirmed postnatally with initially depressed renal function might undergo complete resolution within several years and few months, respectively. However, later-onset urinary tract, metabolic, and neurodevelopmental features of this mutation might appear over years. Therefore, genetic molecular evaluation/diagnosis and continuous follow-up for evolving features are mandatory in affected children.
Collapse
Affiliation(s)
- Roxana Cleper
- Pediatric Nephrology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Adi Reches
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Prenatal Genetic Diagnosis Unit, Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Dana Shapira
- Pediatric Nephrology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sharon Simchoni
- Prenatal Genetic Diagnosis Unit, Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Lewis Reisman
- Pediatric Nephrology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Liat Ben-Sira
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Pediatric Radiology Unit, Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Yuval Yaron
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Prenatal Genetic Diagnosis Unit, Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Igal Wolman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Ultrasound Unit in Obstetrics and Gynecology, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Gustavo Malinger
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Ultrasound Unit in Obstetrics and Gynecology, Lis Maternity Hospital, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Dana Brabbing-Goldstein
- Prenatal Genetic Diagnosis Unit, Genetic Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shay Ben-Shachar
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
50
|
An infant with hyperechoic cystic kidneys and congenital diaphragmatic hernia: Answers. Pediatr Nephrol 2021; 36:4085-4087. [PMID: 34355258 DOI: 10.1007/s00467-021-05155-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 05/17/2021] [Indexed: 10/20/2022]
|