1
|
Sun Q, Xu P, Mao A, Huang S, Li J, Chen L, Li J, Kan H, Huang J, Ji W, Si D, Yan J, Chen ZJ, Gao X, Gao Y. Targeted long-read sequencing enables higher diagnostic yield of ADPKD by accurate PKD1 genetic analysis. NPJ Genom Med 2025; 10:22. [PMID: 40069205 PMCID: PMC11897170 DOI: 10.1038/s41525-025-00477-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 02/03/2025] [Indexed: 03/15/2025] Open
Abstract
Genetic diagnosis of ADPKD has been challenging due to the variant heterogeneity, presence of duplicated segments, and high GC content of exon 1 in PKD1. In our reproductive center, 40 patients were still genetically undiagnosed or diagnosed without single-nucleotide resolution after testing with a short-read sequencing panel in 312 patients with ADPKD phenotype. A combination of long-range PCR and long-read sequencing approach for PKD1 was performed on these 40 patients. LRS additionally identified 10 pathogenic or likely pathogenic PKD1 variants, including four patients with microgene conversion (c.160_166dup, c.2180T>C, and c.8161+1G>A) between PKD1 and its pseudogenes, three with indels (c.-49_43del, c.2985+2_2985+4del, and c.10709_10760dup), one with likely pathogenic deep intronic variant (c.2908-107G>A) and two with large deletions. LRS also identified nine PKD1 CNVs and precisely determined the breakpoints, while SRS failed to identify two of these CNVs. Therefore, LRS enables higher diagnostic yield of ADPKD and provides significant benefits for genetic counseling.
Collapse
Affiliation(s)
- Qian Sun
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Peiwen Xu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Aiping Mao
- Department of Research and Development, Berry Genomics Corporation, Beijing, China
| | - Sexin Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Jie Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Libao Chen
- Department of Research and Development, Berry Genomics Corporation, Beijing, China
| | - Jing Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Haopeng Kan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Ju Huang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Wenkai Ji
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Dayong Si
- School of Life Science, Jilin University, Changchun, China
| | - Junhao Yan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
| | - Zi-Jiang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China
- Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
- Department of Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xuan Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China.
| | - Yuan Gao
- State Key Laboratory of Reproductive Medicine and Offspring Health, Center for Reproductive Medicine, Institute of Women, Children and Reproductive Health, Cheeloo College of Medicine, Shandong University, Jinan, China.
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Shandong University, Jinan, China.
- Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China.
- Shandong Technology Innovation Center for Reproductive Health, Jinan, China.
- Shandong Provincial Clinical Research Center for Reproductive Health, Jinan, China.
- Shandong Key Laboratory of Reproductive Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
- Research Unit of Gametogenesis and Health of ART-Offspring, Chinese Academy of Medical Sciences (No.2021RU001), Jinan, China.
| |
Collapse
|
2
|
Ozyavuz Cubuk P, Akin Duman T. New Variants Identified by Next-Generation Sequencing in Polycystic Kidney Disease Patients. Biochem Genet 2024; 62:5144-5156. [PMID: 38971859 DOI: 10.1007/s10528-024-10880-9] [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: 02/18/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Polycystic kidney disease (PKD) is a common inherited disease characterized by multiple cysts in kidneys and various extra renal manifestations. Molecular diagnosis plays a crucial role in confirming both the clinical diagnosis and preimplantation genetic diagnosis furthermore, selecting appropriate treatment options. This study aimed to expand the understanding of genetic mutations in patients with polycystic kidney disease and to improve the management of patients. The study included 92 patients with a clinical diagnosis of PKD based on renal ultrasound criteria. Targeted next-generation sequencing was performed using a custom panel kit. Of the 92 patients included in the study, pathogenic/likely pathogenic variants of the PKD1, PKD2 genes were detected in 37 patients (40.2%), while 8 patients (8.6%) had variants with uncertain clinical significance. After the additional assessment of pathogenic/likely pathogenic variants, it was found that 15 of the variants in PKD1 and 2 of the variants in PKD2 have not been reported in the literature previously. Additionally, pathogenic variants, 5 of which were novel, have been identified in different genes in 8 patients. This study presented the largest patient cohort conducted in Turkey. These findings were significant in expanding our understanding of the genetic variations associated with polycystic kidney disease. The study contrıbuted the literature data on polycystic kidney disease by reporting important findings that could pave the way for further investigations in the diagnosis, treatment, and management of the affected patients.
Collapse
Affiliation(s)
- Pelin Ozyavuz Cubuk
- Department of Medical Genetics, Haseki Training and Research Hospital, Health Sciences University, Fatih/Istanbul, Turkey.
| | - Tugba Akin Duman
- Department of Medical Genetics, Haseki Training and Research Hospital, Health Sciences University, Fatih/Istanbul, Turkey
| |
Collapse
|
3
|
Rai V, Singh M, Holthoff JH. New Mutation Associated with Polycystic Kidney Disease Type I: A Case Report. Genes (Basel) 2024; 15:1262. [PMID: 39457385 PMCID: PMC11507877 DOI: 10.3390/genes15101262] [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: 08/26/2024] [Revised: 09/17/2024] [Accepted: 09/25/2024] [Indexed: 10/28/2024] Open
Abstract
INTRODUCTION Autosomal dominant polycystic kidney disease (ADPKD) is one of the most prevalent heritable disorders, characterized by the progressive development of kidney cysts leading to renal failure. It is primarily caused by mutations in the PKD1 and PKD2 genes, which account for approximately 85% and 15% of cases, respectively. This case report describes a previously unreported mutation in the PKD1 gene, identified in a family involving an aunt and her niece with ADPKD. CASE PRESENTATION The index case, a 56-year-old female with chronic kidney disease stage 3b secondary to ADPKD and hypertension, exhibited a strong family history of polycystic kidney disease (PKD). Initial genetic evaluations did not identify any recognized pathogenic mutations, leading to a more detailed investigation which revealed a novel mutation in the PKD1 gene. This mutation was also found in her niece, who presented with early-onset disease. CONCLUSIONS The identification of a heterozygous six-nucleotide deletion, c.2084_2089del, resulting in the in-frame deletion of two amino acids, p.Pro695_Ala696del, in the PKD1 gene, has been linked with ADPKD in these patients. This report emphasizes the need for continuous updates to genetic data for a deeper understanding of the diagnosis and prognosis of ADPKD that could potentially aid in targeted therapy.
Collapse
Affiliation(s)
- Vanya Rai
- Mayo Clinic, 200 First Street SW, Rochester, MN 55905, USA;
| | - Manisha Singh
- Department of Nephrology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| | - Joseph H. Holthoff
- Department of Nephrology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA;
| |
Collapse
|
4
|
Palomero OE, DeCaen PG. ADPKD variants in the PKD2 pore helix cause structural collapse of the gate and distinct forms of channel dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612744. [PMID: 39314384 PMCID: PMC11419077 DOI: 10.1101/2024.09.12.612744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
PKD2 is a member of the polycystin subfamily of transient receptor potential (TRP) ion channel subunits which traffic and function in primary cilia organelle membranes. Millions of individuals carry pathogenic genetic variants in PKD2 that cause a life-threatening condition called autosomal dominant polycystic kidney disease (ADPKD). Although ADPKD is a common monogenetic disorder, there is no drug cure or available therapeutics which address the underlying channel dysregulation. Furthermore, the structural and mechanistic impact of most disease-causing variants are uncharacterized. Using direct cilia electrophysiology, cryogenic electron microscopy (cryo-EM), and super resolution imaging, we have discovered mechanistic differences in channel dysregulation caused by three germline missense variants located in PKD2's pore helix 1. Variant C632R reduces protein thermal stability, resulting in impaired channel assembly and abolishes primary cilia trafficking. In contrast, variants F629S and R638C retain native cilia trafficking, but exhibit gating defects. Resolved cryo-EM structures (2.7-3.2Å) of the variants indicate loss of critical pore helix interactions and precipitate allosteric collapse of the channels inner gate. Results demonstrate how ADPKD-causing these mutations have divergent and ranging impacts on PKD2 function, despite their shared structural proximity. These unexpected findings underscore the need for mechanistic characterization of polycystin variants, which may guide rational drug development of ADPKD therapeutics. Regarding polycystin nomenclature The revised and current IUPHAR/BPS nomenclature creates ambiguity regarding the genetic identity of the polycystin family members of transient receptor potential ion channels (TRPP), especially when cross-referencing manuscripts that describe subunits using the former system 1 . Traditionally, the products of polycystin genes (e.g., PKD2) are referred to as polycystin proteins (e.g., polycystin-2). For simplicity and to prevent confusion, we will refer to the polycystin gene name rather than differentiating gene and protein with separate names- a nomenclature we have recently outlined (Annual Reviews in Physiology, Esarte Palomero et al. 2023) 2.
Collapse
|
5
|
Achkar KA, Abdelnour LM, Abu Jawdeh BG, Tantisattamoa E, Al Ammary F. Evaluation and Long-Term Follow-Up of Living Kidney Donors. ADVANCES IN KIDNEY DISEASE AND HEALTH 2024; 31:400-407. [PMID: 39232610 DOI: 10.1053/j.akdh.2024.04.003] [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: 10/01/2023] [Revised: 03/21/2024] [Accepted: 04/10/2024] [Indexed: 09/06/2024]
Abstract
The evaluation of living kidney donor candidates is a complex and lengthy process. Donor candidates face geographic and socioeconomic barriers to completing donor evaluation. Inequities in access to living donations persist. With a growing demand for kidney transplants and a shortage of living donors, transplant centers are more permissive of accepting less-than-ideal donor candidates. Donors have an increased lifetime risk of kidney failure, but the absolute risk increase is small. Efforts are needed to support donor candidates to complete donor nephrectomy safely and efficiently and receive optimal follow-up care to prevent risk factors for kidney disease and detect complications early. In this article, the authors address key elements of donor kidney evaluation, including current living donation policy requirements and transplant center practices. The authors present a simplified comprehensive practical approach to help guide providers in completing donor evaluation and follow-up care with best outcomes possible.
Collapse
Affiliation(s)
| | - Lama M Abdelnour
- Department of Medicine, University of California Los Angeles, Los Angeles, CA
| | | | | | - Fawaz Al Ammary
- Department of Medicine, University of California Irvine, Orange, CA.
| |
Collapse
|
6
|
Zhang Y, Yu C, Li X. Kidney Aging and Chronic Kidney Disease. Int J Mol Sci 2024; 25:6585. [PMID: 38928291 PMCID: PMC11204319 DOI: 10.3390/ijms25126585] [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: 04/27/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
The process of aging inevitably leads to an increase in age-related comorbidities, including chronic kidney disease (CKD). In many aspects, CKD can be considered a state of accelerated and premature aging. Aging kidney and CKD have numerous common characteristic features, ranging from pathological presentation and clinical manifestation to underlying mechanisms. The shared mechanisms underlying the process of kidney aging and the development of CKD include the increase in cellular senescence, the decrease in autophagy, mitochondrial dysfunction, and the alterations of epigenetic regulation, suggesting the existence of potential therapeutic targets that are applicable to both conditions. In this review, we provide a comprehensive overview of the common characteristics between aging kidney and CKD, encompassing morphological changes, functional alterations, and recent advancements in understanding the underlying mechanisms. Moreover, we discuss potential therapeutic strategies for targeting senescent cells in both the aging process and CKD.
Collapse
Affiliation(s)
- Yingying Zhang
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Chen Yu
- Department of Nephrology, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai 200092, China;
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
7
|
Xu D, Mao A, Chen L, Wu L, Ma Y, Mei C. Comprehensive Analysis of PKD1 and PKD2 by Long-Read Sequencing in Autosomal Dominant Polycystic Kidney Disease. Clin Chem 2024; 70:841-854. [PMID: 38527221 DOI: 10.1093/clinchem/hvae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 01/23/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by heterogeneous variants in the PKD1 and PKD2 genes. Genetic analysis of PKD1 has been challenging due to homology with 6 PKD1 pseudogenes and high GC content. METHODS A single-tube multiplex long-range-PCR and long-read sequencing-based assay termed "comprehensive analysis of ADPKD" (CAPKD) was developed and evaluated in 170 unrelated patients by comparing to control methods including next-generation sequencing (NGS) and multiplex ligation-dependent probe amplification. RESULTS CAPKD achieved highly specific analysis of PKD1 with a residual noise ratio of 0.05% for the 6 pseudogenes combined. CAPKD identified PKD1 and PKD2 variants (ranging from variants of uncertain significance to pathogenic) in 160 out of the 170 patients, including 151 single-nucleotide variants (SNVs) and insertion-deletion variants (indels), 6 large deletions, and one large duplication. Compared to NGS, CAPKD additionally identified 2 PKD1 variants (c.78_96dup and c.10729_10732dup). Overall, CAPKD increased the rate of variant detection from 92.9% (158/170) to 94.1% (160/170), and the rate of diagnosis with pathogenic or likely pathogenic variants from 82.4% (140/170) to 83.5% (142/170). CAPKD also directly determined the cis-/trans-configurations in 11 samples with 2 or 3 SNVs/indels, and the breakpoints of 6 large deletions and one large duplication, including 2 breakpoints in the intron 21 AG-repeat of PKD1, which could only be correctly characterized by aligning to T2T-CHM13. CONCLUSIONS CAPKD represents a comprehensive and specific assay toward full characterization of PKD1 and PKD2 variants, and improves the genetic diagnosis for ADPKD.
Collapse
Affiliation(s)
- Dechao Xu
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Aiping Mao
- Department of Third-Generation Sequencing, Berry Genomics Corporation, Beijing, China
| | - Libao Chen
- Department of Third-Generation Sequencing, Berry Genomics Corporation, Beijing, China
| | - Le Wu
- Department of Third-Generation Sequencing, Berry Genomics Corporation, Beijing, China
| | - Yiyi Ma
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Changlin Mei
- Department of Nephrology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
8
|
Yen PW, Chen YA, Wang W, Mao FS, Chao CT, Chiang CK, Lin SH, Tarng DC, Chiu YW, Wu MJ, Chen YC, Kao JTW, Wu MS, Lin CL, Huang JW, Hung KY. The screening, diagnosis, and management of patients with autosomal dominant polycystic kidney disease: A national consensus statement from Taiwan. Nephrology (Carlton) 2024; 29:245-258. [PMID: 38462235 DOI: 10.1111/nep.14287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 03/12/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of end-stage kidney disease (ESKD) worldwide. Guidelines for the diagnosis and management of ADPKD in Taiwan remains unavailable. In this consensus statement, we summarize updated information on clinical features of international and domestic patients with ADPKD, followed by suggestions for optimal diagnosis and care in Taiwan. Specifically, counselling for at-risk minors and reproductive issues can be important, including ethical dilemmas surrounding prenatal diagnosis and pre-implantation genetic diagnosis. Studies reveal that ADPKD typically remains asymptomatic until the fourth decade of life, with symptoms resulting from cystic expansion with visceral compression, or rupture. The diagnosis can be made based on a detailed family history, followed by imaging studies (ultrasound, computed tomography, or magnetic resonance imaging). Genetic testing is reserved for atypical cases mostly. Common tools for prognosis prediction include total kidney volume, Mayo classification and PROPKD/genetic score. Screening and management of complications such as hypertension, proteinuria, urological infections, intracranial aneurysms, are also crucial for improving outcome. We suggest that the optimal management strategies of patients with ADPKD include general medical care, dietary recommendations and ADPKD-specific treatments. Key points include rigorous blood pressure control, dietary sodium restriction and Tolvaptan use, whereas the evidence for somatostatin analogues and mammalian target of rapamycin (mTOR) inhibitors remains limited. In summary, we outline an individualized care plan emphasizing careful monitoring of disease progression and highlight the need for shared decision-making among these patients.
Collapse
Affiliation(s)
- Pao-Wen Yen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Yung-An Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Wei Wang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Fang-Sheng Mao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chia-Ter Chao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan City, Taiwan
| | - Chih-Kang Chiang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Yung-Chang Chen
- Kidney Research Center, Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Juliana Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Mai-Szu Wu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Liang Lin
- Division of Nephrology, Department of Internal Medicine, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi County, Taiwan
| | - Jenq-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Kuan-Yu Hung
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| |
Collapse
|
9
|
Wei T, Zhang B, Tang W, Li X, Shuai Z, Tang T, Zhang Y, Deng L, Liu Q. A de novo PKD1 mutation in a Chinese family with autosomal dominant polycystic kidney disease. Medicine (Baltimore) 2024; 103:e27853. [PMID: 38552045 PMCID: PMC10977567 DOI: 10.1097/md.0000000000027853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 11/02/2021] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND PKD1, which has a relatively high mutation rate, is highly polymorphic, and the role of PKD1 is incompletely defined. In the current study, in order to determine the molecular etiology of a family with autosomal dominant polycystic kidney disease, the pathogenicity of an frameshift mutation in the PKD1 gene, c.9484delC, was evaluated. METHODS The family clinical data were collected. Whole exome sequencing analysis determined the level of this mutation in the proband's PKD1, and Sanger sequencing and bioinformatics analysis were performed. SIFT, Polyphen2, and MutationTaster were used to evaluate the conservation of the gene and pathogenicity of the identified mutations. SWISS-MODEL was used to predict and map the protein structure of PKD1 and mutant neonate proteins. RESULTS A novel c.9484delC (p.Arg3162Alafs*154) mutation of the PKD1 gene was identified by whole exome sequencing in the proband, which was confirmed by Sanger sequencing in his sister (II7). The same mutation was not detected in the healthy pedigree members. Random screening of 100 normal and end-stage renal disease patients did not identify the c.9484delC mutation. Bioinformatics analysis suggested that the mutation caused the 3162 nd amino acid substitution of arginine by alanine and a shift in the termination codon. As a result, the protein sequence was shortened from 4302 amino acids to 3314 amino acids, the protein structure was greatly changed, and the PLAT/LH2 domain was destroyed. Clustal analysis indicated that the altered amino acids were highly conserved in mammals. CONCLUSION A novel mutation in the PKD1 gene has been identified in an affected Chinese family. The mutation is probably responsible for a range of clinical manifestations for which reliable prenatal diagnosis and genetic counseling may be provided.
Collapse
Affiliation(s)
- Ting Wei
- Department of Medical Laboratory, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
- Department of Medical Laboratory, North Sichuan Medical College, Nanchong, China
| | - Bing Zhang
- Department of Medical Laboratory, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Wei Tang
- Department of Medical Laboratory, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Xin Li
- Department of Medical Laboratory, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, China
| | - Zhuang Shuai
- Department of Cardiology Medicine, the Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Tao Tang
- Department of Medical Laboratory, North Sichuan Medical College, Nanchong, China
| | - Yueyang Zhang
- Department of Medical Laboratory, North Sichuan Medical College, Nanchong, China
| | - Lin Deng
- Department of Medical Laboratory, North Sichuan Medical College, Nanchong, China
| | - Qingsong Liu
- Department of Prenatal Diagnosis, Chengdu Women’s and Children’s Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
10
|
Chen PL, Chen CF, Lin HYH, Riley DJ, Chen Y. The Link between Autosomal Dominant Polycystic Kidney Disease and Chromosomal Instability: Exploring the Relationship. Int J Mol Sci 2024; 25:2936. [PMID: 38474184 PMCID: PMC10932443 DOI: 10.3390/ijms25052936] [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: 01/29/2024] [Revised: 02/27/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD) with germline mutations in a PKD1 or PKD2 gene, innumerable cysts develop from tubules, and renal function deteriorates. Second-hit somatic mutations and renal tubular epithelial (RTE) cell death are crucial features of cyst initiation and disease progression. Here, we use established RTE lines and primary ADPKD cells with disease-associated PKD1 mutations to investigate genomic instability and DNA damage responses. We found that ADPKD cells suffer severe chromosome breakage, aneuploidy, heightened susceptibility to DNA damage, and delayed checkpoint activation. Immunohistochemical analyses of human kidneys corroborated observations in cultured cells. DNA damage sensors (ATM/ATR) were activated but did not localize at nuclear sites of damaged DNA and did not properly activate downstream transducers (CHK1/CHK2). ADPKD cells also had the ability to transform, as they achieved high saturation density and formed colonies in soft agar. Our studies indicate that defective DNA damage repair pathways and the somatic mutagenesis they cause contribute fundamentally to the pathogenesis of ADPKD. Acquired mutations may alternatively confer proliferative advantages to the clonally expanded cell populations or lead to apoptosis. Further understanding of the molecular details of aberrant DNA damage responses in ADPKD is ongoing and holds promise for targeted therapies.
Collapse
Affiliation(s)
- Phang-Lang Chen
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA; (P.-L.C.); (C.-F.C.)
| | - Chi-Fen Chen
- Department of Biological Chemistry, University of California, Irvine, CA 92697, USA; (P.-L.C.); (C.-F.C.)
| | - Hugo Y.-H. Lin
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Daniel J. Riley
- Department of Medicine, Division of Nephrology, University of Texas Health, San Antonio, TX 78245, USA;
| | - Yumay Chen
- Department of Medicine, Division of Endocrinology, University of California, Irvine, CA 92697, USA
| |
Collapse
|
11
|
Nakashima M, Suga N, Ikeda Y, Yoshikawa S, Matsuda S. Inspiring Tactics with the Improvement of Mitophagy and Redox Balance for the Development of Innovative Treatment against Polycystic Kidney Disease. Biomolecules 2024; 14:207. [PMID: 38397444 PMCID: PMC10886467 DOI: 10.3390/biom14020207] [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: 12/21/2023] [Revised: 01/31/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Polycystic kidney disease (PKD) is the most common genetic form of chronic kidney disease (CKD), and it involves the development of multiple kidney cysts. Not enough medical breakthroughs have been made against PKD, a condition which features regional hypoxia and activation of the hypoxia-inducible factor (HIF) pathway. The following pathology of CKD can severely instigate kidney damage and/or renal failure. Significant evidence verifies an imperative role for mitophagy in normal kidney physiology and the pathology of CKD and/or PKD. Mitophagy serves as important component of mitochondrial quality control by removing impaired/dysfunctional mitochondria from the cell to warrant redox homeostasis and sustain cell viability. Interestingly, treatment with the peroxisome proliferator-activated receptor-α (PPAR-α) agonist could reduce the pathology of PDK and might improve the renal function of the disease via the modulation of mitophagy, as well as the condition of gut microbiome. Suitable modulation of mitophagy might be a favorable tactic for the prevention and/or treatment of kidney diseases such as PKD and CKD.
Collapse
Affiliation(s)
| | | | | | | | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women’s University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
12
|
Zhao Q, Tan Y, Xiao X, Xiang Q, Yang M, Wang H, Liu S. A novel heterozygous PKD1 variant causing alternative splicing in a Chinese family with autosomal dominant polycystic kidney disease. Mol Genet Genomic Med 2023; 11:e2217. [PMID: 37272738 PMCID: PMC10422069 DOI: 10.1002/mgg3.2217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 06/06/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by pathogenic variants of PKD1 and PKD2. Compared to PKD2-related patients, patients with PKD1 pathogenic variants have more severe symptoms, present a gradual decline in renal function, and finally progress to end-stage kidney disease with an earlier mean onset age. METHODS In this study, trio exome sequencing (ES) was performed to reveal the genetic etiology in a Chinese family clinically diagnosed with polycystic kidney, followed by validation through Sanger sequencing on both genomic DNA and cDNA levels. Subsequently, targeted preimplantation genetic testing was provided for the family. RESULTS A novel heterozygous PKD1 variant (c.1717_1722+11del) was detected in the proband and other clinically-affected relatives. Interestingly, cDNA sequencing demonstrated that the variant, despite being annotated as non-frameshift within exon 8, impacted the splicing of PKD1. Two abnormal transcription products were formed: one induced frameshift, while the other caused 133 amino acids to be inserted between exon 8 and exon 9. CONCLUSIONS Our study revealed a novel PKD1 variant using ES as the cause of ADPKD in a Chinese family with multiple affected members. The variant at the exon-intron boundary would induce alternative splicing, which should not be excluded from genetic analysis. Validated on the cDNA level could provide more comprehensive genetic information for disease stratification. And the novel variant expands the spectrum of PKD1 variants in ADPKD. The recurrent risk could be blocked accordingly for the families' offspring.
Collapse
Affiliation(s)
- Qianying Zhao
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Yu Tan
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Xiao Xiao
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Qinqin Xiang
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Mei Yang
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - He Wang
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| | - Shanling Liu
- Department of Medical GeneticsWest China Second University Hospital, Sichuan UniversityChengduChina
- Department of Obstetrics and GynecologyWest China Second University Hospital, Sichuan UniversityChengduChina
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of EducationChengduChina
| |
Collapse
|
13
|
Pei XM, Yeung MHY, Wong ANN, Tsang HF, Yu ACS, Yim AKY, Wong SCC. Targeted Sequencing Approach and Its Clinical Applications for the Molecular Diagnosis of Human Diseases. Cells 2023; 12:493. [PMID: 36766834 PMCID: PMC9913990 DOI: 10.3390/cells12030493] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/19/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023] Open
Abstract
The outbreak of COVID-19 has positively impacted the NGS market recently. Targeted sequencing (TS) has become an important routine technique in both clinical and research settings, with advantages including high confidence and accuracy, a reasonable turnaround time, relatively low cost, and fewer data burdens with the level of bioinformatics or computational demand. Since there are no clear consensus guidelines on the wide range of next-generation sequencing (NGS) platforms and techniques, there is a vital need for researchers and clinicians to develop efficient approaches, especially for the molecular diagnosis of diseases in the emergency of the disease and the global pandemic outbreak of COVID-19. In this review, we aim to summarize different methods of TS, demonstrate parameters for TS assay designs, illustrate different TS panels, discuss their limitations, and present the challenges of TS concerning their clinical application for the molecular diagnosis of human diseases.
Collapse
Affiliation(s)
- Xiao Meng Pei
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Martin Ho Yin Yeung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Alex Ngai Nick Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong 999077, China
| | - Hin Fung Tsang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong 999077, China
- Department of Clinical Laboratory and Pathology, Hong Kong Adventist Hospital, Hong Kong, China
| | - Allen Chi Shing Yu
- Codex Genetics Limited, Unit 212, 2/F., Building 16W, No. 16 Science Park West Avenue, The Hong Kong Science Park, Hong Kong 852, China
| | - Aldrin Kay Yuen Yim
- Codex Genetics Limited, Unit 212, 2/F., Building 16W, No. 16 Science Park West Avenue, The Hong Kong Science Park, Hong Kong 852, China
| | - Sze Chuen Cesar Wong
- Department of Applied Biology & Chemical Technology, The Hong Kong Polytechnic University, Hong Kong 999077, China
| |
Collapse
|
14
|
Wu R, Bai B, Li F, Bai R, Zhuo Y, Zhu Z, Jia R, Li S, Chen Y, Lan X. Phenotypes and genetic etiology of spontaneous polycystic kidney and liver disease in cynomolgus monkey. Front Vet Sci 2023; 10:1106016. [PMID: 36876010 PMCID: PMC9978152 DOI: 10.3389/fvets.2023.1106016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 01/25/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Polycystic kidney disease (PKD) is a common autosomal dominant or recessive genetic disease, often accompanied by polycystic liver disease (PLD). Many cases of PKD in animals have been reported. However, little is known about the genes that cause PKD in animals. Methods In this study, we evaluated the clinical phenotypes of PKD in two spontaneously aged cynomolgus monkeys and explored the genetic etiology using whole-genome sequencing (WGS). Ultrasonic and histological consequences were further investigated in PKD- and PLD-affected monkeys. Results The results indicated that the kidneys of the two monkeys had varying degrees of cystic changes, and the renal cortex was thinned and accompanied by fluid accumulation. As for hepatopathy, inflammatory cell infiltration, cystic effusion, steatosis of hepatocytes, and pseudo-lobular were found. Based on WGS results, the variants of PKD1:(XM_015442355: c.1144G>C p. E382Q) and GANAB: (NM_001285075.1: c.2708T>C/p. V903A) are predicted to be likely pathogenic heterozygous mutations in PKD- and PLD-affected monkeys. Discussion Our study suggests that the cynomolgus monkey PKD and PLD phenotypes are very similar to those in humans, and are probably caused by pathogenic genes homologous to humans. The results indicate that cynomolgus monkeys can be used as the most appropriate animal model for human PKD pathogenesis research and therapeutic drug screening.
Collapse
Affiliation(s)
- Ruo Wu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Bing Bai
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Feng Li
- Kunming Biomed International, Kunming, China
| | - Raoxian Bai
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Yan Zhuo
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Zhengna Zhu
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Rongfang Jia
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Shangang Li
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Yongchang Chen
- State Key Laboratory of Primate Biomedical Research and Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, China.,Yunnan Key Laboratory of Primate Biomedical Research, Kunming, China
| | - Xiaoping Lan
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Hosseinpour M, Ardalani F, Mohseni M, Beheshtian M, Arzhangi S, Ossareh S, Najmabadi H, Nobakht A, Kahrizi K, Broumand B. Targeted Next Generation Sequencing Revealed Novel Variants in the PKD1 and PKD2 Genes of Iranian Patients with Autosomal Dominant Polycystic Kidney Disease. ARCHIVES OF IRANIAN MEDICINE 2022; 25:600-608. [PMID: 37543885 PMCID: PMC10685772 DOI: 10.34172/aim.2022.95] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/10/2022] [Indexed: 08/07/2023]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD), one of the common inherited disorders in humans, is characterized by the development and enlargement of renal cysts, often leading to end-stage renal disease (ESRD). In this study, Iranian ADPKD families were subjected to high-throughput DNA sequencing to find potential causative variants facilitating the way toward risk assessment and targeted therapy. METHODS Our protocol was based on the targeted next generation sequencing (NGS) panel previously developed in our center comprising 12 genes involved in PKD. This panel has been applied to investigate the genetic causes of 32 patients with a clinical suspicion of ADPKD. RESULTS We identified a total of 31 variants for 32 individuals, two of which were each detected in two individuals. Twenty-seven out of 31 detected variants were interpreted as pathogenic/likely pathogenic and the remaining 4 of uncertain significance with a molecular diagnostic success rate of 87.5%. Among these variants, 25 PKD1/2 pathogenic/likely pathogenic variants were detected in 32 index patients (78.1%), and variants of uncertain significance in four individuals (12.5% in PKD1/2). The majority of variants was identified in PKD1 (74.2%). Autosomal recessive PKD was identified in one patient, indicating the similarities between recessive and dominant PKD. In concordance with earlier studies, this biallelic PKD1 variant, p.Arg3277Cys, leads to rapidly progressive and severe disease with very early-onset ADPKD. CONCLUSION Our findings suggest that targeted gene panel sequencing is expected to be the method of choice to improve diagnostic and prognostic accuracy in PKD patients with heterogeneity in genetic background.
Collapse
Affiliation(s)
- Maryam Hosseinpour
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Fariba Ardalani
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Marzieh Mohseni
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Maryam Beheshtian
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Sanaz Arzhangi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Shahrzad Ossareh
- Division of Nephrology, Department of Medicine, Hasheminejad Kidney Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Ali Nobakht
- Department of Nephrology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Behrooz Broumand
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Tehran, Iran
| |
Collapse
|
16
|
Xu J, Xue C, Wang X, Zhang L, Mei C, Mao Z. Chromatin Methylation Abnormalities in Autosomal Dominant Polycystic Kidney Disease. Front Med (Lausanne) 2022; 9:921631. [PMID: 35865176 PMCID: PMC9294145 DOI: 10.3389/fmed.2022.921631] [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: 04/16/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease worldwide and is one of the major causes of end-stage renal disease. PKD1 and PKD2 are two genes that mainly contribute to the development and progression of ADPKD. The precise mechanism is not fully understood. In recent years, epigenetic modification has drawn increasing attention. Chromatin methylation is a very important category of PKD epigenetic changes and mostly involves DNA, histone, and RNA methylation. Genome hypomethylation and regional gene hypermethylation coexist in ADPKD. We found that the genomic DNA of ADPKD kidney tissues showed extensive demethylation by whole-genome bisulphite sequencing, while some regional DNA methylation from body fluids, such as blood and urine, can be used as diagnostic or prognostic biomarkers to predict PKD progression. Histone modifications construct the histone code mediated by histone methyltransferases and contribute to aberrant methylation changes in PKD. Considering the complexity of methylation abnormalities occurring in different regions and genes on the PKD epigenome, more specific therapy aiming to restore to the normal genome should lead to the development of epigenetic treatment.
Collapse
Affiliation(s)
- Jing Xu
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Cheng Xue
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Xiaodong Wang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
| | - Lei Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, China
- Lei Zhang
| | - Changlin Mei
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
- Changlin Mei
| | - Zhiguo Mao
- Kidney Institute, Department of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, China
- *Correspondence: Zhiguo Mao
| |
Collapse
|
17
|
Watanabe M, Umeyama K, Nakano K, Matsunari H, Fukuda T, Matsumoto K, Tajiri S, Yamanaka S, Hasegawa K, Okamoto K, Uchikura A, Takayanagi S, Nagaya M, Yokoo T, Nakauchi H, Nagashima H. Generation of heterozygous PKD1 mutant pigs exhibiting early-onset renal cyst formation. J Transl Med 2022; 102:560-569. [PMID: 34980882 PMCID: PMC9042704 DOI: 10.1038/s41374-021-00717-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 11/16/2021] [Accepted: 11/27/2021] [Indexed: 11/08/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, manifesting as the progressive development of fluid-filled renal cysts. In approximately half of all patients with ADPKD, end-stage renal disease results in decreased renal function. In this study, we used CRISPR-Cas9 and somatic cell cloning to produce pigs with the unique mutation c.152_153insG (PKD1insG/+). Pathological analysis of founder cloned animals and progeny revealed that PKD1insG/+ pigs developed many pathological conditions similar to those of patients with heterozygous mutations in PKD1. Pathological similarities included the formation of macroscopic renal cysts at the neonatal stage, number and cystogenic dynamics of the renal cysts formed, interstitial fibrosis of the renal tissue, and presence of a premature asymptomatic stage. Our findings demonstrate that PKD1insG/+ pigs recapitulate the characteristic symptoms of ADPKD.
Collapse
Affiliation(s)
- Masahito Watanabe
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Kazuhiro Umeyama
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Kazuaki Nakano
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Hitomi Matsunari
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Toru Fukuda
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Kei Matsumoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Susumu Tajiri
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Shuichiro Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Koki Hasegawa
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Kazutoshi Okamoto
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Ayuko Uchikura
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Shuko Takayanagi
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Masaki Nagaya
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Distinguished Professor Unit, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo, 108-8639, Japan
- Institute for Stem Cell Biology and Regenerative Medicine, Department of Genetics, Stanford University School of Medicine, 265 Campus Drive, Stanford, CA, 94305, USA
| | - Hiroshi Nagashima
- Meiji University International Institute for Bio-Resource Research, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan.
- Laboratory of Medical Bioengineering, Department of Life Sciences, School of Agriculture, Meiji University, 1-1-1 Higashimita, Tama-ku, Kawasaki, Kanagawa, 214-8571, Japan.
| |
Collapse
|
18
|
Keith KA, Reed LK, Nguyen A, Qaiser R. Neurovascular Syndromes. Neurosurg Clin N Am 2021; 33:135-148. [PMID: 34801137 DOI: 10.1016/j.nec.2021.09.010] [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: 10/19/2022]
Abstract
Patients with cerebrovascular syndromes are at risk for additional concerns associated with their syndrome. A wide variety of syndromes are associated with cerebrovascular diseases. Multidisciplinary care is helpful to ensure comprehensive evaluation and management. Precise diagnosis and appreciation for the underlying syndrome is critical for effective cerebrovascular and broader care. This text focuses on these conditions with a focus on underlying pathophysiology and associated genetics, presentation, diagnosis, and management of each disease.
Collapse
Affiliation(s)
- Kristin A Keith
- Baylor Scott & White Health/Texas A&M Neurosurgery Department, 2401 South 31st Street, MS-01-610A, Temple, TX 76508, USA
| | - Laura K Reed
- Baylor Scott & White Health/Texas A&M Neurosurgery Department, 2401 South 31st Street, MS-01-610A, Temple, TX 76508, USA
| | - Anthony Nguyen
- Baylor Scott & White Health/Texas A&M Neurosurgery Department, 2401 South 31st Street, MS-01-610A, Temple, TX 76508, USA
| | - Rabia Qaiser
- Baylor Scott & White Health/Texas A&M Neurosurgery Department, 2401 South 31st Street, MS-01-610A, Temple, TX 76508, USA.
| |
Collapse
|
19
|
Nephroplex: a kidney-focused NGS panel highlights the challenges of PKD1 sequencing and identifies a founder BBS4 mutation. J Nephrol 2021; 34:1855-1874. [PMID: 33964006 PMCID: PMC8610957 DOI: 10.1007/s40620-021-01048-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 04/04/2021] [Indexed: 12/02/2022]
Abstract
Background Genetic testing of patients with inherited kidney diseases has emerged as a tool of clinical utility by improving the patients’ diagnosis, prognosis, surveillance and therapy. Methods The present study applied a Next Generation Sequencing (NGS)-based panel, named NephroPlex, testing 115 genes causing renal diseases, to 119 individuals, including 107 probands and 12 relatives. Thirty-five (poly)cystic and 72 non (poly)cystic individuals were enrolled. The latter subgroup of patients included Bardet-Biedl syndrome (BBS) patients, as major components. Results Disease-causing mutations were identified in 51.5 and 40% of polycystic and non-polycystic individuals, respectively. Autosomal dominant polycystic kidney disease (ADPKD) patients with truncating PKD1 variants showed a trend towards a greater slope of the age-estimated glomerular filtration rate (eGFR) regression line than patients with (i) missense variants, (ii) any PKD2 mutations and (iii) no detected mutations, according to previous findings. The analysis of BBS individuals showed a similar frequency of BBS4,9,10 and 12 mutations. Of note, all BBS4-mutated patients harbored the novel c.332+1G>GTT variant, which was absent in public databases, however, in our internal database, an additional heterozygote carrier was found. All BBS4-mutated individuals originated from the same geographical area encompassing the coastal provinces of Naples. Discussion In conclusion, these findings indicate the potential for a genetic panel to provide useful information at both clinical and epidemiological levels. Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1007/s40620-021-01048-4.
Collapse
|
20
|
Zamani M, Seifi T, Sedighzadeh S, Negahdari S, Zeighami J, Sedaghat A, Yadegari T, Saberi A, Hamid M, Shariati G, Galehdari H. Whole-Exome Sequencing Application for Genetic Diagnosis of Kidney Diseases: A Study from Southwest of Iran. KIDNEY360 2021; 2:873-877. [PMID: 35373060 PMCID: PMC8791347 DOI: 10.34067/kid.0006902020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 03/10/2021] [Indexed: 02/04/2023]
Affiliation(s)
- Mina Zamani
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran,Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Tahereh Seifi
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran,Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Sahar Sedighzadeh
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran,Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Samira Negahdari
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Jawaher Zeighami
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Alireza Sedaghat
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran,Health Research Institute, Diabetes Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Tahereh Yadegari
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran
| | - Alihossein Saberi
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran,Department of Medical Genetics, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Hamid
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran,Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Gholamreza Shariati
- Whole Exome Sequencing Division, Narges Medical Genetics and Prenatal Diagnosis Laboratory, Ahvaz, Iran,Department of Medical Genetics, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hamid Galehdari
- Department of Biology, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
21
|
Australia and New Zealand renal gene panel testing in routine clinical practice of 542 families. NPJ Genom Med 2021; 6:20. [PMID: 33664247 PMCID: PMC7933190 DOI: 10.1038/s41525-021-00184-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 01/25/2021] [Indexed: 12/11/2022] Open
Abstract
Genetic testing in nephrology clinical practice has moved rapidly from a rare specialized test to routine practice both in pediatric and adult nephrology. However, clear information pertaining to the likely outcome of testing is still missing. Here we describe the experience of the accredited Australia and New Zealand Renal Gene Panels clinical service, reporting on sequencing for 552 individuals from 542 families with suspected kidney disease in Australia and New Zealand. An increasing number of referrals have been processed since service inception with an overall diagnostic rate of 35%. The likelihood of identifying a causative variant varies according to both age at referral and gene panel. Although results from high throughput genetic testing have been primarily for diagnostic purposes, they will increasingly play an important role in directing treatment, genetic counseling, and family planning.
Collapse
|
22
|
Ghenu IM, Constantin R, Ionescu D, Dragos D. Giant Cavernous Hemangioma of the Liver in a Patient with Autosomal Dominant Polycystic Kidney Disease. AMERICAN JOURNAL OF CASE REPORTS 2020; 21:e927188. [PMID: 33206631 PMCID: PMC7681259 DOI: 10.12659/ajcr.927188] [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] [Indexed: 11/19/2022]
Abstract
Patient: Male, 41-year-old Final Diagnosis: Autosomal dominant polycystic kidney disease Symptoms: Pain Medication: — Clinical Procedure: Computed tomography • ultrasonography Specialty: Gastroenterology and Hepatology • Medicine, General and Internal • Nephrology
Collapse
Affiliation(s)
- Iuliana Maria Ghenu
- Department of Pharmacology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania.,Department of Oncology, University Emergency Hospital, Bucharest, Romania
| | - Rodica Constantin
- Nephrology Clinic, University Emergency Hospital, Bucharest, Romania
| | - Dorin Ionescu
- Nephrology Clinic, University Emergency Hospital, Bucharest, Romania.,Department of Medical Semiology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Dorin Dragos
- Nephrology Clinic, University Emergency Hospital, Bucharest, Romania.,Department of Medical Semiology, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
23
|
Haumann S, Müller RU, Liebau MC. Metabolic Changes in Polycystic Kidney Disease as a Potential Target for Systemic Treatment. Int J Mol Sci 2020; 21:ijms21176093. [PMID: 32847032 PMCID: PMC7503958 DOI: 10.3390/ijms21176093] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 12/16/2022] Open
Abstract
Autosomal recessive and autosomal dominant polycystic kidney disease (ARPKD, ADPKD) are systemic disorders with pronounced hepatorenal phenotypes. While the main underlying genetic causes of both ARPKD and ADPKD have been well-known for years, the exact molecular mechanisms resulting in the observed clinical phenotypes in the different organs, remain incompletely understood. Recent research has identified cellular metabolic changes in PKD. These findings are of major relevance as there may be an immediate translation into clinical trials and potentially clinical practice. Here, we review important results in the field regarding metabolic changes in PKD and their modulation as a potential target of systemic treatment.
Collapse
Affiliation(s)
- Sophie Haumann
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany;
| | - Roman-Ulrich Müller
- Department II of Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany;
- CECAD, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Systems Biology of Ageing Cologne, University of Cologne, 50931 Cologne, Germany
| | - Max C. Liebau
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50937 Cologne, Germany;
- Center for Molecular Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, 50931 Cologne, Germany
- Correspondence: ; Tel.: +49-221-478-4359
| |
Collapse
|
24
|
Kim H, Kim HH, Chang CL, Song SH, Kim N. Novel PKD1 Mutations in Patients with Autosomal Dominant Polycystic Kidney Disease. Lab Med 2020; 52:174-180. [PMID: 32816041 DOI: 10.1093/labmed/lmaa047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVE Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disease. Identifying mutated causative genes can provide diagnostic and prognostic information. In this study, we describe the clinical application of a next generation sequencing (NGS)-based, targeted multi-gene panel test for the genetic diagnosis of patients with ADPKD. METHODS We applied genetic analysis on 26 unrelated known or suspected patients with ADPKD. A total of 10 genes related to cystic change of kidney were targeted. Detected variants were classified according to standard guidelines. RESULTS We identified 19 variants (detection rate: 73.1%), including PKD1 (n = 18) and PKD2 (n = 1). Of the 18 PKD1 variants, 8 were novel. CONCLUSION Multigene panel test can be a comprehensive tool in a clinical setting for genetic diagnosis of ADPKD. It allows us to identify clinically significant novel variants and confirm the diagnosis, and these objectives are difficult to achieve using conventional diagnostic tools.
Collapse
Affiliation(s)
- Hyerin Kim
- Department of Laboratory Medicine, Pusan National University Hospital, Busan, Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Hyung-Hoi Kim
- Department of Laboratory Medicine, Pusan National University Hospital, Busan, Korea.,Biomedical Research Institute, Pusan National University Hospital, Busan, Korea
| | - Chulhun L Chang
- Department of Laboratory Medicine, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Sang Heon Song
- Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.,Division of Nephrology, Department of Internal Medicine, Pusan National University Hospital, Busan, Korea
| | - Namhee Kim
- Biomedical Research Institute, Pusan National University Hospital, Busan, Korea.,Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, Korea
| |
Collapse
|
25
|
Ta CM, Vien TN, Ng LCT, DeCaen PG. Structure and function of polycystin channels in primary cilia. Cell Signal 2020; 72:109626. [PMID: 32251715 DOI: 10.1016/j.cellsig.2020.109626] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 12/12/2022]
Abstract
Variants in genes which encode for polycystin-1 and polycystin-2 cause most forms of autosomal dominant polycystic disease (ADPKD). Despite our strong understanding of the genetic determinants of ADPKD, we do not understand the structural features which govern the function of polycystins at the molecular level, nor do we understand the impact of most disease-causing variants on the conformational state of these proteins. These questions have remained elusive because polycystins localize to several organelle membranes, including the primary cilia. Primary cilia are microtubule based organelles which function as cellular antennae. Polycystin-2 and related polycystin-2 L1 are members of the transient receptor potential (TRP) ion channel family, and form distinct ion channels in the primary cilia of disparate cell types which can be directly measured. Polycystin-1 has both ion channel and adhesion G-protein coupled receptor (GPCR) features-but its role in forming a channel complex or as a channel subunit chaperone is undetermined. Nonetheless, recent polycystin structural determination by cryo-EM has provided a molecular template to understand their biophysical regulation and the impact of disease-causing variants. We will review these advances and discuss hypotheses regarding the regulation of polycystin channel opening by their structural domains within the context of the primary cilia.
Collapse
Affiliation(s)
- Chau My Ta
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA
| | - Thuy N Vien
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA
| | - Leo C T Ng
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA
| | - Paul G DeCaen
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, 320 E Superior, Chicago, IL 60611, USA.
| |
Collapse
|
26
|
Tsukiyama T, Kobayashi K, Nakaya M, Iwatani C, Seita Y, Tsuchiya H, Matsushita J, Kitajima K, Kawamoto I, Nakagawa T, Fukuda K, Iwakiri T, Izumi H, Itagaki I, Kume S, Maegawa H, Nishinakamura R, Nishio S, Nakamura S, Kawauchi A, Ema M. Monkeys mutant for PKD1 recapitulate human autosomal dominant polycystic kidney disease. Nat Commun 2019; 10:5517. [PMID: 31822676 PMCID: PMC6904451 DOI: 10.1038/s41467-019-13398-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/07/2019] [Indexed: 12/16/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) caused by PKD1 mutations is one of the most common hereditary disorders. However, the key pathological processes underlying cyst development and exacerbation in pre-symptomatic stages remain unknown, because rodent models do not recapitulate critical disease phenotypes, including disease onset in heterozygotes. Here, using CRISPR/Cas9, we generate ADPKD models with PKD1 mutations in cynomolgus monkeys. As in humans and mice, near-complete PKD1 depletion induces severe cyst formation mainly in collecting ducts. Importantly, unlike in mice, PKD1 heterozygote monkeys exhibit cyst formation perinatally in distal tubules, possibly reflecting the initial pathology in humans. Many monkeys in these models survive after cyst formation, and cysts progress with age. Furthermore, we succeed in generating selective heterozygous mutations using allele-specific targeting. We propose that our models elucidate the onset and progression of ADPKD, which will serve as a critical basis for establishing new therapeutic strategies, including drug treatments.
Collapse
Affiliation(s)
- Tomoyuki Tsukiyama
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, 606-8501, Japan.
| | - Kenichi Kobayashi
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
- Department of Urology, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Masataka Nakaya
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, 606-8501, Japan
| | - Chizuru Iwatani
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Yasunari Seita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Hideaki Tsuchiya
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Jun Matsushita
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Kahoru Kitajima
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Ikuo Kawamoto
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Takahiro Nakagawa
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Koji Fukuda
- Shin Nippon Biomedical Laboratories, Ltd, Kagoshima, 891-1394, Japan
| | - Teppei Iwakiri
- Shin Nippon Biomedical Laboratories, Ltd, Kagoshima, 891-1394, Japan
| | - Hiroyuki Izumi
- Shin Nippon Biomedical Laboratories, Ltd, Kagoshima, 891-1394, Japan
| | - Iori Itagaki
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
- The Corporation for Production and Research of Laboratory Primates, Ibaraki, 305-0003, Japan
| | - Shinji Kume
- Department of Medicine, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Hiroshi Maegawa
- Department of Medicine, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Saori Nishio
- Division of Rheumatology, Endocrinology and Nephrology, Hokkaido University Graduate School of Medicine, Hokkaido, 060-8648, Japan
| | - Shinichiro Nakamura
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Akihiro Kawauchi
- Department of Urology, Shiga University of Medical Science, Shiga, 520-2192, Japan
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Shiga, 520-2192, Japan.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, 606-8501, Japan.
- PRESTO, Japan Science and Technology Agency, Saitama, 332-0012, Japan.
| |
Collapse
|
27
|
Chang MY, C M Ong A. Targeting new cellular disease pathways in autosomal dominant polycystic kidney disease. Nephrol Dial Transplant 2019; 33:1310-1316. [PMID: 28992279 DOI: 10.1093/ndt/gfx262] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 07/24/2017] [Indexed: 12/22/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of end-stage renal failure. Understanding the molecular and cellular pathogenesis of ADPKD could help to identify new targets for treatment. The classic cellular cystic phenotype includes changes in proliferation, apoptosis, fluid secretion, extracellular matrix and cilia function. However, recent research, suggests that the cellular cystic phenotype could be broader and that changes, such as altered metabolism, autophagy, inflammation, oxidative stress and epigenetic modification, could play important roles in the processes of cyst initiation, cyst growth or disease progression. Here we review these newer cellular pathways, describe evidence for their possible links to cystic pathogenesis or different stages of disease and discuss the options for developing novel treatments.
Collapse
Affiliation(s)
- Ming-Yang Chang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Albert C M Ong
- Kidney Genetics Group, Academic Nephrology Unit, University of Sheffield Medical School, Sheffield, UK.,Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
28
|
Cornec-Le Gall E, Blais JD, Irazabal MV, Devuyst O, Gansevoort RT, Perrone RD, Chapman AB, Czerwiec FS, Ouyang J, Heyer CM, Senum SR, Le Meur Y, Torres VE, Harris PC. Can we further enrich autosomal dominant polycystic kidney disease clinical trials for rapidly progressive patients? Application of the PROPKD score in the TEMPO trial. Nephrol Dial Transplant 2019; 33:645-652. [PMID: 28992127 PMCID: PMC5888998 DOI: 10.1093/ndt/gfx188] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/13/2017] [Indexed: 01/22/2023] Open
Abstract
Background The PROPKD score has been proposed to stratify the risk of progression to end-stage renal disease in autosomal dominant polycystic kidney disease (ADPKD) subjects. We aimed to assess its prognostic value in a genotyped subgroup of subjects from the Tolvaptan Phase 3 Efficacy and Safety Study in Autosomal Dominant Polycystic Kidney Disease (TEMPO3/4) trial. Methods In the post hoc analysis, PKD1 and PKD2 were screened in 770 subjects and the PROPKD score was calculated in mutation-positive subjects (male: 1 point; hypertension <35 years: 2 points; first urologic event <35 years: 2 points; nontruncating PKD1 mutation: 2 points; truncating PKD1 mutation: 4 points). Subjects were classified into low-risk (LR; 0-3 points), intermediate-risk (IR; 4-6 points) and high-risk (HR; 7-9 points) groups. Results The PROPKD score was calculated in 749 subjects (LR = 132, IR = 344 and HR = 273); age was inversely related to risk (LR = 43.6 years, IR = 39.5 years, HR = 36.2 years; P < 0.001). Subjects from the HR group had significantly higher height-adjusted total kidney volume (TKV) and rates of TKV growth. While baseline renal function was similar across all risk groups, the rate of estimated glomerular filtration rate (eGFR) decline significantly increased from LR to HR in the placebo group. Tolvaptan treatment effectiveness to reduce TKV growth was similar in all three risk categories. While tolvaptan significantly slowed eGFR decline in the IR (tolvaptan = -2.34 versus placebo = -3.33 mL/min/1.73 m2/year; P = 0.008) and HR groups (tolvaptan = -2.74 versus placebo = -3.94 mL/min/1.73 m2/year; P = 0.002), there was no difference in the LR group (tolvaptan = -2.35 versus placebo = -2.50 mL/min/1.73 m2/year; P = 0.72). Excluding the LR subjects from the analysis improved the apparent treatment effect of tolvaptan on eGFR decline. Conclusion This study confirms the prognostic value of the PROPKD score and suggests that it could reduce costs and enhance endpoint sensitivity by enriching future study populations for rapidly progressing ADPKD subjects.
Collapse
Affiliation(s)
- Emilie Cornec-Le Gall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA.,European University of Western Brittany, CHU Brest, Brest, France
| | | | - Maria V Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA
| | - Olivier Devuyst
- Institute of Nephrology, University of Zurich, Zurich, Switzerland
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center of Groningen, Groningen, The Netherlands
| | | | | | | | | | - Christina M Heyer
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA
| | - Sarah R Senum
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA
| | - Yannick Le Meur
- European University of Western Brittany, CHU Brest, Brest, France
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
29
|
Siwy J, Mischak H, Zürbig P. Proteomics and personalized medicine: a focus on kidney disease. Expert Rev Proteomics 2019; 16:773-782. [DOI: 10.1080/14789450.2019.1659138] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Justyna Siwy
- R & D, Mosaiques Diagnostics GmbH, Hannover, Germany
| | - Harald Mischak
- R & D, Mosaiques Diagnostics GmbH, Hannover, Germany
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Petra Zürbig
- R & D, Mosaiques Diagnostics GmbH, Hannover, Germany
| |
Collapse
|
30
|
Dotto RP, de Santana LS, Lindsey SC, Caetano LA, Franco LF, Moisés RCMS, Sa JR, Nishiura JL, Teles MG, Heilberg IP, Dias-da-Silva MR, Giuffrida FMA, Reis AF. Searching for mutations in the HNF1B gene in a Brazilian cohort with renal cysts and hyperglycemia. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:250-257. [PMID: 31066763 PMCID: PMC10522195 DOI: 10.20945/2359-3997000000138] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 02/13/2019] [Indexed: 01/27/2023]
Abstract
OBJECTIVE To verify the presence of variants in HNF1B in a sample of the Brazilian population selected according to the presence of renal cysts associated with hyperglycemia. SUBJECTS AND METHODS We evaluated 28 unrelated patients with clinical suspicion of HNF1B mutation because of the concomitant presence of diabetes mellitus (DM) or prediabetes and renal cysts. Genotyping was accomplished using Sanger sequencing or multiplex ligation-dependent probe amplification (MLPA). In positive cases, available relatives were recruited. RESULTS We found two patients with HNF1B mutations. The first presented the variant p.Pro328Leufs*48(c.983delC) and had DM, renal cysts, and hypomagnesemia. The second presented a heterozygous whole gene deletion in HNF1B, DM, renal cysts, body and tail pancreatic agenesis, and hypomagnesemia; this alteration was also found in his two siblings and his father. CONCLUSION The recruitment of suspected cases of HNF1B gene mutations in Brazilians due to hyperglycemia and renal cysts presents two positive cases. Our cases contribute to the annotation of clinical and biochemical phenotypes of this rare form of maturity-onset diabetes of the young (MODY).
Collapse
Affiliation(s)
- Renata P. Dotto
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Lucas Santos de Santana
- Universidade de São PauloUnidade de Endocrinologia Genética/LIM25Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilGrupo de Diabetes Monogênico, Unidade de Endocrinologia Genética/LIM25, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil
| | - Susan C. Lindsey
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Lilian Araújo Caetano
- Universidade de São PauloUnidade de Endocrinologia Genética/LIM25Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilGrupo de Diabetes Monogênico, Unidade de Endocrinologia Genética/LIM25, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil
| | - Luciana F. Franco
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Regina Célia M. S. Moisés
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - João R. Sa
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - José Luiz Nishiura
- Universidade Federal de São PauloDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Milena Gurgel Teles
- Universidade de São PauloUnidade de Endocrinologia Genética/LIM25Faculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilGrupo de Diabetes Monogênico, Unidade de Endocrinologia Genética/LIM25, Faculdade de Medicina, Universidade de São Paulo (FMUSP), São Paulo, SP, Brasil
| | - Ita P. Heilberg
- Universidade Federal de São PauloDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Magnus R. Dias-da-Silva
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| | - Fernando M. A. Giuffrida
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
- Universidade do Estado da BahiaDepartamento de Ciências da VidaUniversidade do Estado da BahiaSalvadorBABrasilDepartamento de Ciências da Vida, Universidade do Estado da Bahia (UNEB), Salvador, BA, Brasil
| | - André F. Reis
- Universidade Federal de São PauloDisciplina de EndocrinologiaDepartamento de MedicinaUniversidade Federal de São PauloSão PauloSPBrasilDisciplina de Endocrinologia, Departamento de Medicina, Universidade Federal de São Paulo (Unifesp), São Paulo, SP, Brasil
| |
Collapse
|
31
|
Kim JH, Park EY, Chitayat D, Stachura DL, Schaper J, Lindstrom K, Jewett T, Wieczorek D, Draaisma JM, Sinnema M, Hoeberigs C, Hempel M, Bachman KK, Seeley AH, Stone JK, Kong HK, Vukadin L, Richard A, Shinde DN, McWalter K, Si YC, Douglas G, Lim ST, Vissers LELM, Lemaire M, Ahn EYE. SON haploinsufficiency causes impaired pre-mRNA splicing of CAKUT genes and heterogeneous renal phenotypes. Kidney Int 2019; 95:1494-1504. [PMID: 31005274 DOI: 10.1016/j.kint.2019.01.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 12/10/2018] [Accepted: 01/04/2019] [Indexed: 10/27/2022]
Abstract
Although genetic testing is increasingly used in clinical nephrology, a large number of patients with congenital abnormalities of the kidney and urinary tract (CAKUT) remain undiagnosed with current gene panels. Therefore, careful curation of novel genetic findings is key to improving diagnostic yields. We recently described a novel intellectual disability syndrome caused by de novo heterozygous loss-of-function mutations in the gene encoding the splicing factor SON. Here, we show that many of these patients, including two previously unreported, exhibit a wide array of kidney abnormalities. Detailed phenotyping of 14 patients with SON haploinsufficiency identified kidney anomalies in 8 patients, including horseshoe kidney, unilateral renal hypoplasia, and renal cysts. Recurrent urinary tract infections, electrolyte disturbances, and hypertension were also observed in some patients. SON knockdown in kidney cell lines leads to abnormal pre-mRNA splicing, resulting in decreased expression of several established CAKUT genes. Furthermore, these molecular events were observed in patient-derived cells with SON haploinsufficiency. Taken together, our data suggest that the wide spectrum of phenotypes in patients with a pathogenic SON mutation is a consequence of impaired pre-mRNA splicing of several CAKUT genes. We propose that genetic testing panels designed to diagnose children with a kidney phenotype should include the SON gene.
Collapse
Affiliation(s)
- Jung-Hyun Kim
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Eun Young Park
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - David Chitayat
- Division of Clinical and Metabolic Genetics, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada; The Prenatal Diagnosis and Medical Genetics Program, Department of Obstetrics and Gynecology, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - David L Stachura
- Department of Biological Sciences, California State University Chico, Chico, California, USA
| | - Jörg Schaper
- Institute of Diagnostic and Interventional Radiology, University of Düsseldorf, Düsseldorf, Germany
| | - Kristin Lindstrom
- Division of Genetics and Metabolism, Phoenix Children's Hospital, Phoenix, Arizona, USA
| | - Tamison Jewett
- Department of Pediatrics, Section on Medical Genetics, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Dagmar Wieczorek
- Institute of Human Genetics, University Clinic Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany; Institute of Human Genetics, University Clinic Essen, University Duisburg-Essen, Essen, Germany
| | - Jos M Draaisma
- Department of Pediatrics, Radboudumc Amalia Children's Hospital, Nijmegen, The Netherlands
| | - Margje Sinnema
- Department of Clinical Genetics and School for Oncology & Developmental Biology (GROW), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Christianne Hoeberigs
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Maja Hempel
- Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | - Joshua K Stone
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Hyun Kyung Kong
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Lana Vukadin
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | - Alexander Richard
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA
| | | | | | | | | | - Ssang-Taek Lim
- Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Lisenka E L M Vissers
- Department of Human Genetics, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mathieu Lemaire
- Division of Nephrology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada; Cell Biology Program, SickKids Research Institute, University of Toronto, Toronto, Ontario, Canada.
| | - Eun-Young Erin Ahn
- Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA; Department of Biochemistry and Molecular Biology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.
| |
Collapse
|
32
|
Ali H, Al-Mulla F, Hussain N, Naim M, Asbeutah AM, AlSahow A, Abu-Farha M, Abubaker J, Al Madhoun A, Ahmad S, Harris PC. PKD1 Duplicated regions limit clinical Utility of Whole Exome Sequencing for Genetic Diagnosis of Autosomal Dominant Polycystic Kidney Disease. Sci Rep 2019; 9:4141. [PMID: 30858458 PMCID: PMC6412018 DOI: 10.1038/s41598-019-40761-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 02/21/2019] [Indexed: 12/18/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited monogenic renal disease characterised by the accumulation of clusters of fluid-filled cysts in the kidneys and is caused by mutations in PKD1 or PKD2 genes. ADPKD genetic diagnosis is complicated by PKD1 pseudogenes located proximal to the original gene with a high degree of homology. The next generation sequencing (NGS) technology including whole exome sequencing (WES) and whole genome sequencing (WGS), is becoming more affordable and its use in the detection of ADPKD mutations for diagnostic and research purposes more widespread. However, how well does NGS technology compare with the Gold standard (Sanger sequencing) in the detection of ADPKD mutations? Is a question that remains to be answered. We have evaluated the efficacy of WES, WGS and targeted enrichment methodologies in detecting ADPKD mutations in the PKD1 and PKD2 genes in patients who were clinically evaluated by ultrasonography and renal function tests. Our results showed that WES detected PKD1 mutations in ADPKD patients with 50% sensitivity, as the reading depth and sequencing quality were low in the duplicated regions of PKD1 (exons 1-32) compared with those of WGS and target enrichment arrays. Our investigation highlights major limitations of WES in ADPKD genetic diagnosis. Enhancing reading depth, quality and sensitivity of WES in the PKD1 duplicated regions (exons 1-32) is crucial for its potential diagnostic or research applications.
Collapse
Affiliation(s)
- Hamad Ali
- Department of Medical Laboratory Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Jabriya, Kuwait.
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait.
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait.
| | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait.
| | - Naser Hussain
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Medhat Naim
- Division of Nephrology, Mubarak Al-Kabeer Hospital, Ministry of Health, Jabriya, Kuwait
| | - Akram M Asbeutah
- Department of Radiological Sciences, Faculty of Allied Health Sciences, Health Sciences Center, Kuwait University, Jabriya, Kuwait
| | - Ali AlSahow
- Division of Nephrology, Al-Jahra Hospital, Ministry of Health, Al-Jahra, Kuwait
| | - Mohamed Abu-Farha
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Jehad Abubaker
- Department of Biochemistry and Molecular Biology, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Ashraf Al Madhoun
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute (DDI), Dasman, Kuwait
| | - Sajjad Ahmad
- Department of Cornea and External Diseases, Moorfields Eye Hospital-NHS Foundation Trust, London, United Kingdom
- Institute of Ophthalmology, University Collage London (UCL), London, United Kingdom
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, USA
| |
Collapse
|
33
|
Tsai YC, Teng IL, Jiang ST, Lee YC, Chiou YY, Cheng FY. Safe Nanocomposite-Mediated Efficient Delivery of MicroRNA Plasmids for Autosomal Dominant Polycystic Kidney Disease (ADPKD) Therapy. Adv Healthc Mater 2019; 8:e1801358. [PMID: 30672150 DOI: 10.1002/adhm.201801358] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/03/2019] [Indexed: 12/14/2022]
Abstract
There is currently no cure for gene mutation-caused autosomal dominant polycystic kidney disease (ADPKD). Over half of patients with ADPKD eventually develop kidney failure, requiring dialysis or kidney transplantation. Current treatment modalities for ADPKD focus on reducing morbidity and mortality from renal and extrarenal complications of the disease. MicroRNA has been shown to be useful in treating ADPKD. This study combines anti-miRNA plasmids and iron oxide/alginate nanoparticles for conjugation with antikidney antibodies. These nanocomposites can specifically target renal tubular cells, providing a potential treatment for ADPKD. Magnetic resonance imaging and in vivo imaging system results show effective targeting of renal cells. Anti-miRNA plasmids released from the nanocomposites inhibit cell proliferation and cyst formation in the PKD cellular and animal models. The results suggest the novel combination of the anti-miRNA plasmids and nanomaterials provides potential clinical implications for ADPKD treatment.
Collapse
Affiliation(s)
- Yen-Chang Tsai
- Institute of Clinical Medicine; Medical College; National Cheng Kung University; Tainan 701 Taiwan
| | - I-Ling Teng
- Division of Pediatric Nephrology; Department of Pediatrics; National Cheng Kung University Hospital; Tainan 704 Taiwan
| | - Si-Tse Jiang
- National Laboratory Animal Center; National Applied Research Laboratories; Tainan 741 Taiwan
| | - Yi-Che Lee
- Division of Nephrology; Department of Internal Medicine; E-DA Hospital; I-Shou University; Kaohsiung 824 Taiwan
| | - Yuan-Yow Chiou
- Institute of Clinical Medicine; Medical College; National Cheng Kung University; Tainan 701 Taiwan
- Division of Pediatric Nephrology; Department of Pediatrics; National Cheng Kung University Hospital; Tainan 704 Taiwan
- Department of Pediatrics; College of Medicine; National Cheng Kung University; Tainan 704 Taiwan
| | - Fong-Yu Cheng
- Department of Chemistry; Chinese Culture University; 55, Hwa-Kang Road, Yang-Ming-Shan Taipei 11114 Taiwan
| |
Collapse
|
34
|
Chumley P, Zhou J, Mrug S, Chacko B, Parant JM, Challa AK, Wilson LS, Berryhill TF, Barnes S, Kesterson RA, Bell PD, Darley-Usmar VM, Yoder BK, Mrug M. Truncating PKHD1 and PKD2 mutations alter energy metabolism. Am J Physiol Renal Physiol 2019; 316:F414-F425. [PMID: 30566001 PMCID: PMC6442375 DOI: 10.1152/ajprenal.00167.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 12/04/2018] [Accepted: 12/11/2018] [Indexed: 02/06/2023] Open
Abstract
Deficiency in polycystin 1 triggers specific changes in energy metabolism. To determine whether defects in other human cystoproteins have similar effects, we studied extracellular acidification and glucose metabolism in human embryonic kidney (HEK-293) cell lines with polycystic kidney and hepatic disease 1 ( PKHD1) and polycystic kidney disease (PKD) 2 ( PKD2) truncating defects along multiple sites of truncating mutations found in patients with autosomal recessive and dominant PKDs. While neither the PKHD1 or PKD2 gene mutations nor their position enhanced cell proliferation rate in our cell line models, truncating mutations in these genes progressively increased overall extracellular acidification over time ( P < 0.001 for PKHD1 and PKD2 mutations). PKHD1 mutations increased nonglycolytic acidification rate (1.19 vs. 1.03, P = 0.002), consistent with an increase in tricarboxylic acid cycle activity or breakdown of intracellular glycogen. In addition, they increased basal and ATP-linked oxygen consumption rates [7.59 vs. 5.42 ( P = 0.015) and 4.55 vs. 2.98 ( P = 0.004)]. The PKHD1 and PKD2 mutations also altered mitochondrial morphology, resembling the effects of polycystin 1 deficiency. Together, these data suggest that defects in major PKD genes trigger changes in mitochondrial energy metabolism. After validation in in vivo models, these initial observations would indicate potential benefits of targeting energy metabolism in the treatment of PKDs.
Collapse
Affiliation(s)
- Phillip Chumley
- Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Juling Zhou
- Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - Sylvie Mrug
- Department of Psychology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Balu Chacko
- Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | - John M Parant
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham , Birmingham, Alabama
| | - Anil K Challa
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Landon S Wilson
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham , Birmingham, Alabama
| | - Taylor F Berryhill
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham , Birmingham, Alabama
| | - Stephen Barnes
- Targeted Metabolomics and Proteomics Laboratory, University of Alabama at Birmingham , Birmingham, Alabama
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham , Birmingham, Alabama
- Department of Genetics, University of Alabama at Birmingham , Birmingham, Alabama
| | - Robert A Kesterson
- Department of Genetics, University of Alabama at Birmingham , Birmingham, Alabama
| | - P Darwin Bell
- Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | | | - Bradley K Yoder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Alabama
| | - Michal Mrug
- Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
- Department of Veterans Affairs Medical Center , Birmingham, Alabama
| |
Collapse
|
35
|
Prasad H, Dang DK, Kondapalli KC, Natarajan N, Cebotaru V, Rao R. NHA2 promotes cyst development in an in vitro model of polycystic kidney disease. J Physiol 2019; 597:499-519. [PMID: 30242840 PMCID: PMC6332743 DOI: 10.1113/jp276796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 08/31/2018] [Indexed: 12/21/2022] Open
Abstract
KEY POINTS Significant and selective up-regulation of the Na+ /H+ exchanger NHA2 (SLC9B2) was observed in cysts of patients with autosomal dominant polycystic kidney disease. Using the MDCK cell model of cystogenesis, it was found that NHA2 increases cyst size. Silencing or pharmacological inhibition of NHA2 inhibits cyst formation in vitro. Polycystin-1 represses NHA2 expression via Ca2+ /NFAT signalling whereas the dominant negative membrane-anchored C-terminal fragment (PC1-MAT) increased NHA2 levels. Drugs (caffeine, theophylline) and hormones (vasopressin, aldosterone) known to exacerbate cysts elicit NHA2 expression. Taken together, the findings reveal NHA2 as a potential new player in salt and water homeostasis in the kidney and in the pathogenesis of polycystic kidney disease. ABSTRACT Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2 encoding polycystin-1 (PC1) and polycystin-2 (PC2), respectively. The molecular pathways linking polycystins to cyst development in ADPKD are still unclear. Intracystic fluid secretion via ion transporters and channels plays a crucial role in cyst expansion in ADPKD. Unexpectedly, we observed significant and selective up-regulation of NHA2, a member of the SLC9B family of Na+ /H+ exchangers, that correlated with cyst size and disease severity in ADPKD patients. Using three-dimensional cultures of MDCK cells to model cystogenesis in vitro, we showed that ectopic expression of NHA2 is causal to increased cyst size. Induction of PC1 in MDCK cells inhibited NHA2 expression with concordant inhibition of Ca2+ influx through store-dependent and -independent pathways, whereas reciprocal activation of Ca2+ influx by the dominant negative membrane-anchored C-terminal tail fragment of PC1 elevated NHA2. We showed that NHA2 is a target of Ca2+ /NFAT signalling and is transcriptionally induced by methylxanthine drugs such as caffeine and theophylline, which are contraindicated in ADPKD patients. Finally, we observed robust induction of NHA2 by vasopressin, which is physiologically consistent with increased levels of circulating vasopressin and up-regulation of vasopressin V2 receptors in ADPKD. Our findings have mechanistic implications on the emerging use of vasopressin V2 receptor antagonists such as tolvaptan as safe and effective therapy for polycystic kidney disease and reveal a potential new regulator of transepithelial salt and water transport in the kidney.
Collapse
Affiliation(s)
- Hari Prasad
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Donna K. Dang
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Kalyan C. Kondapalli
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Niranjana Natarajan
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| | - Valeriu Cebotaru
- Department of MedicineUniversity of Maryland School of MedicineBaltimoreMDUSA
| | - Rajini Rao
- Department of PhysiologyJohns Hopkins University School of MedicineBaltimoreMDUSA
| |
Collapse
|
36
|
Chou LF, Cheng YL, Hsieh CY, Lin CY, Yang HY, Chen YC, Hung CC, Tian YC, Yang CW, Chang MY. Effect of Trehalose Supplementation on Autophagy and Cystogenesis in a Mouse Model of Polycystic Kidney Disease. Nutrients 2018; 11:nu11010042. [PMID: 30585217 PMCID: PMC6356442 DOI: 10.3390/nu11010042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/15/2018] [Accepted: 12/20/2018] [Indexed: 12/28/2022] Open
Abstract
Autophagy impairment has been demonstrated in the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD) and could be a new target of treatment. Trehalose is a natural, nonreducing disaccharide that has been shown to enhance autophagy. Therefore, we investigated whether trehalose treatment reduces renal cyst formation in a Pkd1-hypomorphic mouse model. Pkd1 miRNA transgenic (Pkd1 miR Tg) mice and wild-type littermates were given drinking water supplemented with 2% trehalose from postnatal day 35 to postnatal day 91. The control groups received pure water or 2% sucrose for the control of hyperosmolarity. The effect on kidney weights, cystic indices, renal function, cell proliferation, and autophagic activities was determined. We found that Pkd1 miR Tg mice had a significantly lower renal mRNA expression of autophagy-related genes, including atg5, atg12, ulk1, beclin1, and p62, compared with wild-type control mice. Furthermore, immunohistochemical analysis showed that cystic lining cells had strong positive staining for the p62 protein, indicating impaired degradation of the protein by the autophagy-lysosome pathway. However, trehalose treatment did not improve reduced autophagy activities, nor did it reduce relative kidney weights, plasma blood urea nitrogen levels, or cystatin C levels in Pkd1 miR Tg mice. Histomorphological analysis revealed no significant differences in the renal cyst index, fibrosis score, or proliferative score among trehalose-, sucrose-, and water-treated groups. Our results demonstrate that adding trehalose to drinking water does not modulate autophagy activities and renal cystogenesis in Pkd1-deficient mice, suggesting that an oral supplement of trehalose may not affect the progression of ADPKD.
Collapse
Affiliation(s)
- Li-Fang Chou
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ya-Lien Cheng
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chun-Yih Hsieh
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chan-Yu Lin
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Huang-Yu Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Yung-Chang Chen
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Cheng-Chieh Hung
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ya-Chung Tian
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chih-Wei Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ming-Yang Chang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
37
|
Abstract
Cystic kidneys are common causes of end-stage renal disease, both in children and in adults. Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are cilia-related disorders and the two main forms of monogenic cystic kidney diseases. ADPKD is a common disease that mostly presents in adults, whereas ARPKD is a rarer and often more severe form of polycystic kidney disease (PKD) that usually presents perinatally or in early childhood. Cell biological and clinical research approaches have expanded our knowledge of the pathogenesis of ADPKD and ARPKD and revealed some mechanistic overlap between them. A reduced 'dosage' of PKD proteins is thought to disturb cell homeostasis and converging signalling pathways, such as Ca2+, cAMP, mechanistic target of rapamycin, WNT, vascular endothelial growth factor and Hippo signalling, and could explain the more severe clinical course in some patients with PKD. Genetic diagnosis might benefit families and improve the clinical management of patients, which might be enhanced even further with emerging therapeutic options. However, many important questions about the pathogenesis of PKD remain. In this Primer, we provide an overview of the current knowledge of PKD and its treatment.
Collapse
Affiliation(s)
- Carsten Bergmann
- Department of Medicine, University Hospital Freiburg, Freiburg, Germany.
| | - Lisa M. Guay-Woodford
- Center for Translational Science, Children’s National Health System, Washington, DC, USA
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Dorien J. M. Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
38
|
Xu P, Huang S, Li J, Zou Y, Gao M, Kang R, Yan J, Gao X, Gao Y. A novel splicing mutation in the PKD1 gene causes autosomal dominant polycystic kidney disease in a Chinese family: a case report. BMC MEDICAL GENETICS 2018; 19:198. [PMID: 30424739 PMCID: PMC6234645 DOI: 10.1186/s12881-018-0706-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2018] [Accepted: 10/22/2018] [Indexed: 11/10/2022]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic renal disorder in humans, affecting 1 in 400 to 1000 individuals. Mutations PKD1 (which accounts for 85% of ADPKD and produces polycystin-1) and PKD2 (produces polycystin-2) are responsible for this disease. These two polycystins are critical for maintaining normal renal tubular structures during kidney development. Case presentation We performed genetic analysis on a family with ADPKD. DNA samples extracted from ADPKD patient blood were subject to targeted Next generation sequencing for human a panel of renal disease-related genes. A splicing mutation, c.2854-3C > G (also known as IVS11–3C > G), in the PKD1 gene was found in the 3 patients from the family, but was not found in four unaffected relatives and 100 normal control samples. Reverse transcription-PCR (RT-PCR) was performed to analyse the relative mRNA expression in the patient samples. mRNA sequencing showed that 29 bases inserted into the 3′-end of exon 11 in the PKD1 gene lead to a frameshift mutation. Conclusions The PKD1 c.2854-3C > G mutation leads to a frameshift mutation during translation of the polycystin-1 protein, which eventually led to ADPKD in the Chinese family.
Collapse
Affiliation(s)
- Peiwen Xu
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Sexing Huang
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Jie Li
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Yang Zou
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Ming Gao
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Ranran Kang
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Junhao Yan
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China.,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China.,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China
| | - Xuan Gao
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China. .,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China.
| | - Yuan Gao
- Center for Reproductive Medicine, Shandong University, Jinan, 250001, China. .,National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, 250001, China. .,The Key laboratory for Reproductive Endocrinology of Ministry of Education, Jinan, 250001, China.
| |
Collapse
|
39
|
Soroka S, Alam A, Bevilacqua M, Girard LP, Komenda P, Loertscher R, McFarlane P, Pandeya S, Tam P, Bichet DG. Updated Canadian Expert Consensus on Assessing Risk of Disease Progression and Pharmacological Management of Autosomal Dominant Polycystic Kidney Disease. Can J Kidney Health Dis 2018; 5:2054358118801589. [PMID: 30345064 PMCID: PMC6187423 DOI: 10.1177/2054358118801589] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/22/2018] [Indexed: 12/24/2022] Open
Abstract
PURPOSE The purpose of this article is to update the previously published consensus recommendations from March 2017 discussing the optimal management of adult patients with autosomal dominant polycystic kidney disease (ADPKD). This document focuses on recent developments in genetic testing, renal imaging, assessment of risk regarding disease progression, and pharmacological treatment options for ADPKD. SOURCES OF INFORMATION Published literature was searched in PubMed, the Cochrane Library, and Google Scholar to identify the latest evidence related to the treatment and management of ADPKD. METHODS All pertinent articles were reviewed by the authors to determine if a new recommendation was required, or if the previous recommendation needed updating. The consensus recommendations were developed by the authors based on discussion and review of the evidence. KEY FINDINGS The genetics of ADPKD are becoming more complex with the identification of new and rarer genetic variants such as GANAB. Magnetic resonance imaging (MRI) and computed tomography (CT) continue to be the main imaging modalities used to evaluate ADPKD. Total kidney volume (TKV) continues to be the most validated and most used measure to assess disease progression. Since the publication of the previous consensus recommendations, the use of the Mayo Clinic Classification for prognostication purposes has been validated in patients with class 1 ADPKD. Recent evidence supports the benefits of a low-osmolar diet and dietary sodium restriction in patients with ADPKD. Evidence from the Replicating Evidence of Preserved Renal Function: an Investigation of Tolvaptan Safety and Efficacy in ADPKD (REPRISE) trial supports the use of ADH (antidiuretic hormone) receptor antagonism in patients with ADPKD 18 to 55 years of age with eGFR (estimated glomerular filtration rate) of 25 to 65 mL/min/1.73 m2 or 56 to 65 years of age with eGFR of 25 to 44 mL/min/1.73 m2 with historical evidence of a decline in eGFR >2.0 mL/min/1.73 m2/year. LIMITATIONS Available literature was limited to English language publications and to publications indexed in PubMed, the Cochrane Library, and Google Scholar. IMPLICATIONS Advances in the assessment of the risk of disease progression include the validation of the Mayo Clinic Classification for patients with class 1 ADPKD. Advances in the pharmacological management of ADPKD include the expansion of the use of ADH receptor antagonism in patients 18 to 55 years of age with eGFR of 25 to 65 mL/min/1.73 m2 or 56 to 65 years of age with eGFR of 25 to 44 mL/min/1.73 m2 with historical evidence of a decline in eGFR >2.0 mL/min/1.73 m2/year, as per the results of the REPRISE study.
Collapse
Affiliation(s)
- Steven Soroka
- Division of Nephrology, Dalhousie University, Halifax, NS, Canada
| | - Ahsan Alam
- Division of Nephrology, Royal Victoria Hospital, McGill University, Montréal, QC, Canada
| | - Micheli Bevilacqua
- Division of Nephrology, The University of British Columbia, Vancouver, Canada
| | | | - Paul Komenda
- Division of Nephrology, Seven Oaks General Hospital, University of Manitoba, Winnipeg, Canada
| | - Rolf Loertscher
- Division of Nephrology, Lakeshore General Hospital, McGill University, Pointe-Claire, QC, Canada
| | - Philip McFarlane
- Division of Nephrology, St. Michael’s Hospital, University of Toronto, ON, Canada
| | - Sanjaya Pandeya
- Division of Nephrology, Halton Healthcare, Oakville, ON, Canada
| | - Paul Tam
- Division of Nephrology, Scarborough and Rouge Hospital, ON, Canada
| | - Daniel G. Bichet
- Division of Nephrology, Département de Médecine, Pharmacologie et Physiologie, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, QC, Canada
| |
Collapse
|
40
|
Abdelwahed M, Hilbert P, Ahmed A, Mahfoudh H, Bouomrani S, Dey M, Hachicha J, Kamoun H, Keskes-Ammar L, Belguith N. Mutational analysis in patients with Autosomal Dominant Polycystic Kidney Disease (ADPKD): Identification of five mutations in the PKD1 gene. Gene 2018; 671:28-35. [DOI: 10.1016/j.gene.2018.05.112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/24/2018] [Accepted: 05/30/2018] [Indexed: 01/01/2023]
|
41
|
Pei J, Kinch LN, Grishin NV. FlyXCDB—A Resource for Drosophila Cell Surface and Secreted Proteins and Their Extracellular Domains. J Mol Biol 2018; 430:3353-3411. [DOI: 10.1016/j.jmb.2018.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Revised: 05/31/2018] [Accepted: 06/02/2018] [Indexed: 02/06/2023]
|
42
|
Moisan S, Levon S, Cornec-Le Gall E, Le Meur Y, Audrézet MP, Dostie J, Férec C. Novel long-range regulatory mechanisms controlling PKD2 gene expression. BMC Genomics 2018; 19:515. [PMID: 29986647 PMCID: PMC6038307 DOI: 10.1186/s12864-018-4892-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 06/20/2018] [Indexed: 02/01/2023] Open
Abstract
Background Cis-regulatory elements control gene expression over large distances through the formation of chromatin loops, which allow contact between enhancers and gene promoters. Alterations in cis-acting regulatory systems could be linked to human genetic diseases. Here, we analyse the spatial organization of a large region spanning the polycystic kidney disease 2 (PKD2) gene, one of the genes responsible of autosomal dominant polycystic kidney disease (ADPKD). Results By using chromosome conformation capture carbon copy (5C) technology in primary human renal cyst epithelial cells, we identify novel contacts of the PKD2 promoter with chromatin regions, which display characteristics of regulatory elements. In parallel, by using functional analysis with a reporter assay, we demonstrate that three DNAse I hypersensitive sites regions are involved in the regulation of PKD2 gene expression. Conclusions Finally, through alignment of CCCTC-binding factor (CTCF) sites, we suggest that these novel enhancer elements are brought to the PKD2 promoter by chromatin looping via the recruitment of CTCF. Electronic supplementary material The online version of this article (10.1186/s12864-018-4892-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Stéphanie Moisan
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, Bretagne, France. .,Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, Bretagne, France. .,Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU), Hôpital Morvan, Brest, Bretagne, France.
| | - Stéphanie Levon
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, Bretagne, France.,Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, Bretagne, France
| | - Emilie Cornec-Le Gall
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, Bretagne, France.,Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU), Hôpital Morvan, Brest, Bretagne, France
| | - Yannick Le Meur
- Service de néphrologie, Centre Hospitalier Régional Universitaire (CHRU), Brest, Bretagne, France
| | - Marie-Pierre Audrézet
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, Bretagne, France.,Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU), Hôpital Morvan, Brest, Bretagne, France
| | - Josée Dostie
- Department of Biochemistry and Goodman Cancer Research Center, McGill University, Montréal, Québec, Canada
| | - Claude Férec
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1078, Brest, Bretagne, France. .,Faculté de Médecine et des Sciences de la Santé, Université de Bretagne Occidentale (UBO), Brest, Bretagne, France. .,Laboratoire de Génétique Moléculaire et d'Histocompatibilité, Centre Hospitalier Régional Universitaire (CHRU), Hôpital Morvan, Brest, Bretagne, France. .,Etablissement Français du sang (EFS), Brest, Bretagne, France.
| |
Collapse
|
43
|
Murphy EL, Droher ML, DiMaio MS, Dahl NK. Preimplantation Genetic Diagnosis Counseling in Autosomal Dominant Polycystic Kidney Disease. Am J Kidney Dis 2018; 72:866-872. [PMID: 29606500 DOI: 10.1053/j.ajkd.2018.01.048] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 01/19/2018] [Indexed: 01/09/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common hereditary forms of chronic kidney disease. Mutations within PKD1 or PKD2 lead to innumerable fluid-filled cysts in the kidneys and in some instances, end-stage renal disease (ESRD). Affected individuals have a 50% chance of passing the mutation to each of their offspring. Assisted reproductive technology using preimplantation genetic diagnosis (PGD) allows these individuals to reduce this risk to 1% to 2%. We assess the disease burden of 8 individuals with ADPKD who have undergone genetic testing in preparation for PGD. Clinical features that predict high risk for progression to ESRD in patients with ADPKD include genotype, early onset of hypertension, a urologic event before age 35 years, and a large height-adjusted total kidney volume. Patients may have a family history of intracranial aneurysms or complications involving hepatic cysts, which may further influence the decision to pursue PGD. We also explore the cost, risks, and benefits of using PGD. All patients with ADPKD of childbearing potential, regardless of risk for progression to ESRD or risk for a significant disease burden, will likely benefit from genetic counseling.
Collapse
Affiliation(s)
- Erin L Murphy
- Section of Nephrology, Yale University School of Medicine, North Haven, CT
| | - Madeline L Droher
- Section of Nephrology, Yale University School of Medicine, North Haven, CT
| | - Miriam S DiMaio
- Department of Medical Sciences, Frank H. Netter, MD School of Medicine, Quinnipiac University, North Haven, CT
| | - Neera K Dahl
- Section of Nephrology, Yale University School of Medicine, North Haven, CT.
| |
Collapse
|
44
|
Urinary proteomics using capillary electrophoresis coupled to mass spectrometry for diagnosis and prognosis in kidney diseases. Curr Opin Nephrol Hypertens 2018; 25:494-501. [PMID: 27584928 DOI: 10.1097/mnh.0000000000000278] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE OF REVIEW Urine is the most useful of body fluids for biomarker research. Therefore, we have focused on urinary proteomics, using capillary electrophoresis coupled to mass spectrometry, to investigate kidney diseases in recent years. RECENT FINDINGS Several urinary proteomics studies for the detection of various kidney diseases have indicated the potential of this approach aimed at diagnostic and prognostic assessment. Urinary protein biomarkers such as collagen fragments, serum albumin, α-1-antitrypsin, and uromodulin can help to explain the processes involved during disease progression. SUMMARY Urinary proteomics has been used in several studies in order to identify and validate biomarkers associated with different kidney diseases. These biomarkers, with improved sensitivity and specificity when compared with the current gold standards, provide a significant alternative for diagnosis and prognosis, as well as improving clinical decision-making.
Collapse
|
45
|
Sha YK, Sha YW, Mei LB, Huang XJ, Wang X, Lin SB, Li L, Li P. Use of targeted sequence capture and high-throughput sequencing identifies a novel PKD1 mutation involved in adult polycystic kidney disease. Gene 2017; 634:1-4. [PMID: 28870863 DOI: 10.1016/j.gene.2017.08.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Revised: 08/06/2017] [Accepted: 08/30/2017] [Indexed: 11/24/2022]
Abstract
Polycystic kidney disease (PKD) is a common inherited disease that is characterized by a progressive development of renal cysts. Approximately 85% of PKD cases are due to mutations in the polycystin 1 (PKD1) gene. Here, we report a pedigree containing nine patients with autosomal dominant PKD (ADPKD). Using targeted exome sequencing of PKD1 and PKD2 genes, we identified a novel heterozygous frameshift mutation c.3976_3977insCT (p.F1326Sfs*21) in the PKD1 gene that segregated between affected and unaffected family members. This mutation is currently not present in the 1000 Genomes Project nor ExAC databases and is therefore a novel PKD1 mutation involved in ADPKD. These results provide a novel sequence variant for the genetic analysis of this disease.
Collapse
Affiliation(s)
- Yan-Kun Sha
- Department of Nephrology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou 361003, China
| | - Yan-Wei Sha
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Li-Bin Mei
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Xian-Jing Huang
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Xu Wang
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Shao-Bin Lin
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China
| | - Lin Li
- Central Laboratory, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing 100026, China.
| | - Ping Li
- Department of Reproductive Medicine, Xiamen Maternal and Child Care Hospital, Xiamen 361005, China.
| |
Collapse
|
46
|
Cornec-Le Gall E, Torres VE, Harris PC. Genetic Complexity of Autosomal Dominant Polycystic Kidney and Liver Diseases. J Am Soc Nephrol 2017; 29:13-23. [PMID: 29038287 DOI: 10.1681/asn.2017050483] [Citation(s) in RCA: 239] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Data indicate significant phenotypic and genotypic overlap, plus a common pathogenesis, between two groups of inherited disorders, autosomal dominant polycystic kidney diseases (ADPKD), a significant cause of ESRD, and autosomal dominant polycystic liver diseases (ADPLD), which result in significant PLD with minimal PKD. Eight genes have been associated with ADPKD (PKD1 and PKD2), ADPLD (PRKCSH, SEC63, LRP5, ALG8, and SEC61B), or both (GANAB). Although genetics is only infrequently used for diagnosing these diseases and prognosing the associated outcomes, its value is beginning to be appreciated, and the genomics revolution promises more reliable and less expensive molecular diagnostic tools for these diseases. We therefore propose categorization of patients with a phenotypic and genotypic descriptor that will clarify etiology, provide prognostic information, and better describe atypical cases. In genetically defined cases, the designation would include the disease and gene names, with allelic (truncating/nontruncating) information included for PKD1 Recent data have shown that biallelic disease including at least one weak ADPKD allele is a significant cause of symptomatic, very early onset ADPKD. Including a genic (and allelic) descriptor with the disease name will provide outcome clues, guide treatment, and aid prevalence estimates.
Collapse
Affiliation(s)
- Emilie Cornec-Le Gall
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and.,Department of Nephrology, University Hospital, European University of Brittany, and National Institute of Health and Medical Sciences, INSERM U1078, Brest, France
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota; and
| |
Collapse
|
47
|
Rangan GK, Tchan MC, Tong A, Wong ATY, Nankivell BJ. Recent advances in autosomal-dominant polycystic kidney disease. Intern Med J 2017; 46:883-92. [PMID: 27553994 DOI: 10.1111/imj.13143] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 04/03/2016] [Accepted: 05/08/2016] [Indexed: 12/17/2022]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is the most common genetic renal disease in adults, affecting one in every 1000 Australians. It is caused by loss-of-function heterozygous mutations in either PKD1 or PKD2 , which encode the proteins, polycystin-1 and polycystin-2 respectively. The disease hallmark is the development of hundreds of microscopic fluid-filled cysts in the kidney during early childhood, which grow exponentially and continuously through life at varying rates (between 2% and 10% per year), causing loss of normal renal tissue and up to a 50% lifetime risk of dialysis-dependent kidney failure. Other systemic complications include hypertensive cardiac disease, hepatic cysts, intracranial aneurysms, diverticular disease and hernias. Over the last two decades, advances in the genetics and pathogenesis of this disease have led to novel treatments that reduce the rate of renal cyst growth and may potentially delay the onset of kidney failure. New evidence indicates that conventional therapies (such as angiotensin inhibitors and statins) have mild attenuating effects on renal cyst growth and that systemic levels of vasopressin are critical for promoting renal cyst growth in the postnatal period. Identifying and integrating patient-centred perspectives in clinical trials is also being advocated. This review will provide an update on recent advances in the clinical management of ADPKD.
Collapse
Affiliation(s)
- G K Rangan
- Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia.,Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - M C Tchan
- Department of Genetic Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | - A Tong
- Centre for Kidney Research, The Children's Hospital at Westmead, Sydney, New South Wales, Australia.,Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia
| | - A T Y Wong
- Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - B J Nankivell
- Department of Renal Medicine, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia.,Centre for Transplant and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
48
|
Song X, Haghighi A, Iliuta IA, Pei Y. Molecular diagnosis of autosomal dominant polycystic kidney disease. Expert Rev Mol Diagn 2017; 17:885-895. [PMID: 28724316 DOI: 10.1080/14737159.2017.1358088] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease that accounts for 5-10% of end-stage renal disease in developed countries. Mutations in PKD1 and PKD2 account for a majority of cases. Mutation screening of PKD1 is technically challenging largely due to the complexity resulting from duplication of its first 33 exons in six highly homologous pseudogenes (i.e. PKD1P1-P6). Protocol using locus-specific long-range and nested PCR has enabled comprehensive PKD1 mutation screening but is labor-intensive and costly. Here, the authors review how recent advances in Next Generation Sequencing are poised to transform and extend molecular diagnosis of ADPKD. Areas covered: Key original research articles and reviews of the topic published in English identified through PubMed from 1957-2017. Expert commentary: The authors review current and evolving approaches using targeted resequencing or whole genome sequencing for screening typical as well as challenging cases (e.g. cases with no detectable PKD1 and PKD2 mutations which may be due to somatic mosaicism or other cystic disease; and complex genetics such as bilineal disease).
Collapse
Affiliation(s)
- Xuewen Song
- a Division of Nephrology , University Health Network and University of Toronto , Toronto , ON , Canada
| | - Amirreza Haghighi
- a Division of Nephrology , University Health Network and University of Toronto , Toronto , ON , Canada
| | - Ioan-Andrei Iliuta
- a Division of Nephrology , University Health Network and University of Toronto , Toronto , ON , Canada
| | - York Pei
- a Division of Nephrology , University Health Network and University of Toronto , Toronto , ON , Canada
| |
Collapse
|
49
|
Alkanderi S, Yates LM, Johnson SA, Sayer JA. Lessons learned from a multidisciplinary renal genetics clinic. QJM 2017; 110:453-457. [PMID: 28177086 DOI: 10.1093/qjmed/hcx030] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Inherited renal disorders comprise a significant proportion of cases in both paediatric and adult nephrology services. Genetic advances have advanced rapidly while clinical models of care delivery have remained static. AIM To describe a cohort of patients attending a multidisciplinary renal genetics clinic and the insights gained from this experience. DESIGN AND METHODS A retrospective review of clinic cases and their molecular genetic diagnosis over a 5-year period. RESULTS We report details of 244 individuals including 80 probands who attended the clinic. The commonest reasons for referral was familial haematuria which accounted for 37.5% of cases and cystic kidney disease, accounting for 31% of cases. Eighteen probands had a known molecular genetic diagnosis and were referred for genetic counselling and screening of at risk relatives and management plans. About 62 probands and their families were referred for a precise molecular diagnosis and this was achieved in 26 cases (42%). The most frequent new genetic diagnoses were COL4A5 mutations underlying familial haematuria and familial end stage renal disease. The clinic also allowed for patients with rare renal syndromes to be reviewed, such as ciliopathy syndromes, allowing detailed phenotyping and often a precise molecular genetic diagnosis to be provided. CONCLUSIONS The integration of modern day genetics and genomics into multidisciplinary clinics often allows a precise diagnosis which benefits patients, their relatives and the clinicians providing care and future management.
Collapse
Affiliation(s)
- S Alkanderi
- Newcastle University, Institute of Genetic Medicine, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
| | - L M Yates
- Newcastle University, Institute of Genetic Medicine, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
- Northern Genetics Service, International Centre for Life, Central Parkway, Newcastle NE1 3BZ, UK
| | - S A Johnson
- Great North Children's Hospital, Newcastle upon Tyne, NE1 4LP, UK
| | - J A Sayer
- Newcastle University, Institute of Genetic Medicine, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
- Renal Services, Newcastle upon Tyne NHS Foundation Trust Hospitals, Newcastle upon Tyne NE7 7DN, UK
| |
Collapse
|
50
|
|