58 results on '"Pei, York"'
Search Results
2. Reprogramming of Energy Metabolism in Human PKD1 Polycystic Kidney Disease: A Systems Biology Analysis.
- Author
-
Song X, Pickel L, Sung HK, Scholey J, and Pei Y
- Subjects
- Humans, TRPP Cation Channels metabolism, TRPP Cation Channels genetics, Mitochondria metabolism, Mitochondria genetics, Mechanistic Target of Rapamycin Complex 1 metabolism, Mechanistic Target of Rapamycin Complex 1 genetics, Oxidative Phosphorylation, Gene Expression Regulation, Energy Metabolism, Systems Biology methods, Polycystic Kidney, Autosomal Dominant metabolism, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Multiple alterations of cellular metabolism have been documented in experimental studies of autosomal dominant polycystic kidney disease (ADPKD) and are thought to contribute to its pathogenesis. To elucidate the molecular pathways and transcriptional regulators associated with the metabolic changes of renal cysts in ADPKD, we compared global gene expression data from human PKD1 renal cysts, minimally cystic tissues (MCT) from the same patients, and healthy human kidney cortical tissue samples. We found gene expression profiles of PKD1 renal cysts were consistent with the Warburg effect with gene pathway changes favoring increased cellular glucose uptake and lactate production, instead of pyruvate oxidation. Additionally, mitochondrial energy metabolism was globally depressed, associated with downregulation of gene pathways related to fatty acid oxidation (FAO), branched-chain amino acid (BCAA) degradation, the Krebs cycle, and oxidative phosphorylation (OXPHOS) in renal cysts. Activation of mTORC1 and its two target proto-oncogenes, HIF-1α and MYC, was predicted to drive the expression of multiple genes involved in the observed metabolic reprogramming (e.g., GLUT3 , HK1/HK2 , ALDOA , ENO2 , PKM , LDHA/LDHB , MCT4 , PDHA1 , PDK1/3 , MPC1/2 , CPT2 , BCAT1 , NAMPT ); indeed, their predicted expression patterns were confirmed by our data. Conversely, we found AMPK inhibition was predicted in renal cysts. AMPK inhibition was associated with decreased expression of PGC-1α, a transcriptional coactivator for transcription factors PPARα, ERRα, and ERRγ, all of which play a critical role in regulating oxidative metabolism and mitochondrial biogenesis. These data provide a comprehensive map of metabolic pathway reprogramming in ADPKD and highlight nodes of regulation that may serve as targets for therapeutic intervention.
- Published
- 2024
- Full Text
- View/download PDF
3. Real-life use of tolvaptan in ADPKD: a retrospective analysis of a large Canadian cohort.
- Author
-
Calvaruso L, Yau K, Akbari P, Nasri F, Khowaja S, Wang B, Haghighi A, Khalili K, and Pei Y
- Subjects
- Humans, Tolvaptan therapeutic use, Tolvaptan adverse effects, Retrospective Studies, Antidiuretic Hormone Receptor Antagonists therapeutic use, Antidiuretic Hormone Receptor Antagonists adverse effects, Ontario, Polycystic Kidney, Autosomal Dominant drug therapy, Renal Insufficiency, Chronic drug therapy
- Abstract
Tolvaptan is the first disease-modifying drug proven to slow eGFR decline in high-risk patients with ADPKD. However, barriers from the patient perspective to its use in real-life settings have not been systemically examined in a large cohort. This was a single-center, retrospective study of 523 existing or new patients with ADPKD followed at the Center for Innovative Management of PKD in Toronto, Ontario, between January 1, 2016 to December 31, 2018. All patients underwent clinical assessment including total kidney volume measurements and Mayo Clinic Imaging Class (MCIC). Those who were deemed to be at high risk were offered tolvaptan with their preference (yes or no) and reasons for their choices recorded. Overall, 315/523 (60%) patients had MCIC 1C-1E; however, only 96 (30%) of them were treated with tolvaptan at their last follow-up. Among these high-risk patients, those not treated versus treated with tolvaptan were more likely to have a higher eGFR (82 ± 26 vs. 61 ± 27 ml/min/1.73 m
2 ), CKD stages 1-2 (79% vs. 41%), and MCIC 1C (63% vs. 31%). The most common reasons provided for not taking tolvaptan were lifestyle preference related to the aquaretic effect (51%), older age ≥ 60 (12%), and pregnancy/family planning (6%). In this real-world experience, at least 60% of patients with ADPKD considered to be at high risk for progression to ESKD by imaging were not treated with tolvaptan; most of them had early stages of CKD with well-preserved eGFR and as such, were prime targets for tolvaptan therapy to slow disease progression. Given that the most common reason for tolvaptan refusal was the concern for intolerability of the aquaretic side-effect, strategies to mitigate this may help to reduce this barrier to tolvaptan therapy., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
4. Assessing the Risk of Progression to Kidney Failure in Patients With Autosomal Dominant Polycystic Kidney Disease.
- Author
-
Lanktree MB, Kline T, and Pei Y
- Subjects
- Humans, Adult, TRPP Cation Channels genetics, Mutation, Kidney, Polycystic Kidney, Autosomal Dominant complications, Renal Insufficiency complications
- Abstract
While autosomal dominant polycystic kidney disease (ADPKD) is a dichotomous diagnosis, substantial variability in disease severity exists. Identification of inherited risk through family history, genetic testing, and environmental risk factors through clinical assessment are important components of risk assessment for optimal management of patients with ADPKD. Genetic testing is especially helpful in cases with diagnostic uncertainty, particularly in cases with no apparent family history, in young cases (age less than 25 years) where a definitive diagnosis is sought, or in atypical presentations with early, severe, or discordant findings. Currently, risk assessment in ADPKD may be performed with the use of age-adjusted estimated glomerular filtration rate thresholds, evidence of rapid estimated glomerular filtration rate decline on serial measurements, age- and height-adjusted total kidney volume by Mayo Clinic Imaging Classification, or evidence of early hypertension and urological complications combined with PKD1 or PKD2 mutation class; however, caveats exist with each of these approaches. Fine-tuning of risk stratification with advanced imaging features and biomarkers is the subject of research but is not yet ready for general clinical practice. While conservative treatment strategies will be advised for all patients, those with the greatest rate of disease progression will have the most benefit from aggressive disease-modifying therapy. In this narrative review, we will summarize the evidence behind the clinical assessment and risk stratification of patients with ADPKD., (Copyright © 2023 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
5. Venglustat, a Novel Glucosylceramide Synthase Inhibitor, in Patients at Risk of Rapidly Progressing ADPKD: Primary Results of a Double-Blind, Placebo-Controlled, Phase 2/3 Randomized Clinical Trial.
- Author
-
Gansevoort RT, Hariri A, Minini P, Ahn C, Chapman AB, Horie S, Knebelmann B, Mrug M, Ong ACM, Pei YPC, Torres VE, Modur V, Antonshchuk I, and Perrone RD
- Subjects
- Adult, Humans, Kidney, Glomerular Filtration Rate, Disease Progression, Polycystic Kidney, Autosomal Dominant complications, Renal Insufficiency complications
- Abstract
Rationale & Objective: Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the formation of multiple kidney cysts that leads to growth in total kidney volume (TKV) and progression to kidney failure. Venglustat is a glucosylceramide synthase inhibitor that has been shown to inhibit cyst growth and reduce kidney failure in preclinical models of ADPKD., Study Design: STAGED-PKD was a 2-stage, multicenter, double-blind, randomized, placebo-controlled phase 2/3 study in adults with ADPKD at risk of rapidly progressive disease, who were selected based on Mayo Clinic imaging classification of ADPKD class 1C, 1D, or 1E and an estimated glomerular filtration rate (eGFR) of 30-89.9mL/min/1.73m
2 ., Setting & Participants: Enrollment included 236 and 242 patients in stages 1 and 2, respectively., Interventions: In trial stage 1, the patients were randomized 1:1:1 to venglustat, 8mg; venglustat, 15mg; or placebo. In stage 2, the patients were randomized 1:1 to venglustat, 15mg (highest dose identified as safe and well tolerated in stage 1), or placebo., Outcomes: Primary end points were rate of change in TKV over 18 months in stage 1 and eGFR slope over 24 months in stage 2. Secondary end points were eGFR slope over 18 months (stage 1), rate of change in TKV (stage 2), and safety/tolerability, pain, and fatigue (stages 1 and 2)., Results: A prespecified interim futility analysis showed that venglustat treatment had no effect on the annualized rate of change in TKV over 18 months (stage 1) and had a faster rate of decline in eGFR slope over 24 months (stage 2). Due to this lack of efficacy, the study was terminated early., Limitations: The short follow-up period after the end of treatment and limited generalizability of the findings., Conclusions: In patients with rapidly progressing ADPKD, treatment with venglustat at either 8mg or 15mg showed no change in the rate of change in TKV and a faster rate of eGFR decline in STAGED-PKD despite a dose-dependent decrease in plasma glucosylceramide levels., Funding: This study was funded by Sanofi., Trial Registration: Registered at ClinicalTrials.gov with study number NCT03523728., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
6. Urinary epidermal growth factor/monocyte chemotactic peptide 1 ratio as non-invasive predictor of Mayo clinic imaging classes in autosomal dominant polycystic kidney disease.
- Author
-
Rocchetti MT, Pesce F, Matino S, Piscopo G, di Bari I, Trepiccione F, Capolongo G, Perniola MA, Song X, Khowaja S, Haghighi A, Peters D, Paolicelli S, Pontrelli P, Netti GS, Ranieri E, Capasso G, Moschetta M, Pei Y, and Gesualdo L
- Subjects
- Animals, Humans, Mice, Disease Progression, Epidermal Growth Factor genetics, Kidney, Monocytes pathology, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Background: Age- and height-adjusted total kidney volume is currently considered the best prognosticator in patients with autosomal dominant polycystic kidney disease. We tested the ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 for the prediction of the Mayo Clinic Imaging Classes., Methods: Urinary epidermal growth factor and monocyte chemotactic peptide 1 levels were measured in two independent cohorts (discovery, n = 74 and validation set, n = 177) and healthy controls (n = 59) by immunological assay. Magnetic resonance imaging parameters were used for total kidney volume calculation and the Mayo Clinic Imaging Classification defined slow (1A-1B) and fast progressors (1C-1E). Microarray and quantitative gene expression analysis were used to test epidermal growth factor and monocyte chemotactic peptide 1 gene expression., Results: Baseline ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 correlated with total kidney volume adjusted for height (r = - 0.6, p < 0.001), estimated glomerular filtration rate (r = 0.69 p < 0.001), discriminated between Mayo Clinic Imaging Classes (p < 0.001), and predicted the variation of estimated glomerular filtration rate at 10 years (r = - 0.51, p < 0.001). Conditional Inference Trees identified cut-off levels of the ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 for slow and fast progressors at > 132 (100% slow) and < 25.76 (89% and 86% fast, according to age), with 94% sensitivity and 66% specificity (p = 6.51E-16). Further, the ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 at baseline showed a positive correlation (p = 0.006, r = 0.36) with renal outcome (delta-estimated glomerular filtration rate per year, over a mean follow-up of 4.2 ± 1.2 years). Changes in the urinary epidermal growth factor and monocyte chemotactic peptide 1 were mirrored by gene expression levels in both human kidney cysts (epidermal growth factor: - 5.6-fold, fdr = 0.001; monocyte chemotactic peptide 1: 3.1-fold, fdr = 0.03) and Pkd1 knock-out mouse kidney (Egf: - 14.8-fold, fdr = 2.37E-20, Mcp1: 2.8-fold, fdr = 6.82E-15)., Conclusion: The ratio of urinary epidermal growth factor and monocyte chemotactic peptide 1 is a non-invasive pathophysiological biomarker that can be used for clinical risk stratification in autosomal dominant polycystic kidney disease., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
7. Atypical Polycystic Kidney Disease as defined by Imaging.
- Author
-
Iliuta IA, Win AZ, Lanktree MB, Lee SH, Pourafkari M, Nasri F, Guiard E, Haghighi A, He N, Ingram A, Quist C, Hillier D, Khalili K, and Pei Y
- Subjects
- Humans, TRPP Cation Channels genetics, Kidney pathology, Mutation, Disease Progression, Polycystic Kidney, Autosomal Dominant genetics, Renal Insufficiency, Chronic pathology
- Abstract
Using age- and height-adjusted total kidney volume, the Mayo Clinic Imaging Classification provides a validated approach to assess the risk of chronic kidney disease (CKD) progression in autosomal dominant polycystic kidney disease (ADPKD), but requires excluding patients with atypical imaging patterns, whose clinical characteristics have been poorly defined. We report an analysis of the prevalence, clinical and genetic characteristics of patients with atypical polycystic kidney disease by imaging. Patients from the extended Toronto Genetic Epidemiology Study of Polycystic Kidney Disease recruited between 2016 and 2018 completed a standardized clinical questionnaire, kidney function assessment, genetic testing, and kidney imaging by magnetic resonance or computed tomography. We compared the prevalence, clinical features, genetics, and renal prognosis of atypical versus typical polycystic kidney disease by imaging. Forty-six of the 523 (8.8%) patients displayed atypical polycystic kidney disease by imaging; they were older (55 vs. 43 years; P < 0.001), and less likely to have a family history of ADPKD (26.1% vs. 74.6%; P < 0.001), a detectable PKD1 or PKD2 mutation (9.2% vs. 80.4%; P < 0.001), or progression to CKD stage 3 or stage 5 (P < 0.001). Patients with atypical polycystic kidney disease by imaging represent a distinct prognostic group with a low likelihood of progression to CKD., (© 2022. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
8. Autosomal Dominant Polycystic Kidney Disease: Role of Imaging in Diagnosis and Management.
- Author
-
Odedra D, Sabongui S, Khalili K, Schieda N, Pei Y, and Krishna S
- Subjects
- Humans, Diagnostic Imaging, Kidney, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Polycystic Kidney, Autosomal Dominant therapy, Cysts, Renal Insufficiency
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disorder with progressive renal function decline, and disease severity is determined based on the type of genetic mutation. The diagnosis is usually established at imaging, primarily at US, and is based on age-dependent criteria and the number of visible cysts. ADPKD is classified into class 1 (typical) and class 2 (atypical) according to the Mayo Clinic Imaging Classification (MCIC) system. Height-adjusted total kidney volume (TKV) has emerged as a predictor of future renal function decline and renal failure in ADPKD, and several methods can be used for estimation. MCIC class 1 ADPKD is further subdivided into five types based on height-adjusted TKV (A, B, C, D, and E). Patients with a larger height-adjusted TKV (ie, MCIC 1C-E) are at high risk for progression to end-stage renal disease and will potentially benefit from vasopressin receptor antagonists, which have been shown to reduce the rate of cyst growth and slow renal function decline. Other renal complications primarily relate to hemorrhage within cysts or cyst infections. Subtraction images are key for assessment of complex cysts when malignancy is suspected, as the presence of protein and blood can limit the assessment for an enhancing component. The radiologist has a central role in establishing a diagnosis, excluding mimics, identifying complications, assessing severity, and predicting future renal failure. Interventional radiologists play a therapeutic role in management of complications by cyst drainage, sclerotherapy, or embolization.
© RSNA, 2022 Online supplemental material and the slide presentation from the RSNA Annual Meeting are available for this article.- Published
- 2023
- Full Text
- View/download PDF
9. Safety of High-Dose 3% Sodium Tetradecyl Sulfate for Sclerotherapy of Renal Cysts in Patients with Autosomal Dominant Polycystic Kidney Disease.
- Author
-
Patel NR, Stella SF, Nasser M, Tai E, Hsiao R, Iliuta IA, Jaberi A, Kachura JR, Pei Y, and Shlomovitz E
- Subjects
- Humans, Sclerotherapy adverse effects, Sclerotherapy methods, Sodium Tetradecyl Sulfate adverse effects, United States, Cysts, Polycystic Kidney, Autosomal Dominant drug therapy, Polycystic Kidney, Autosomal Dominant therapy
- Abstract
This study assessed the safety profile of high-volume (>10 mL) 3% sodium tetradecyl sulfate (STS) sclerotherapy for the treatment of renal cysts in patients with autosomal dominant polycystic kidney disease. A total of 211 sclerotherapy treatments were performed in 169 patients over a 5-year period, with a comparison of 2 patient cohorts based on the STS volumes used. The first cohort (n = 112) received a high volume (greater than 10 mL) of STS, and the second cohort (n = 57) received a low volume (less than 10 mL). The minor adverse event rate for the cohorts was 14.5% and 9.6%, respectively (P = .310), with postprocedure pain being the most common event. One major adverse event occurred, for which the patient required hospitalization for infection after low-volume STS treatment. Doses of STS higher than those currently recommended by the Food and Drug Administration for intravascular use allow large renal cysts to be treated safely in the setting of autosomal dominant polycystic kidney disease., (Copyright © 2022 SIR. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
10. Dietary Interventions in Autosomal Dominant Polycystic Kidney Disease.
- Author
-
Pickel L, Iliuta IA, Scholey J, Pei Y, and Sung HK
- Subjects
- Humans, Quality of Life, Disease Progression, Signal Transduction, Caloric Restriction, Kidney, Polycystic Kidney, Autosomal Dominant drug therapy
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the progressive growth of renal cysts, leading to the loss of functional nephrons. Recommendations for individuals with ADPKD to maintain a healthy diet and lifestyle are largely similar to those for the general population. However, recent evidence from preclinical models suggests that more tightly specified dietary regimens, including caloric restriction, intermittent fasting, and ketogenic diets, hold promise to slow disease progression, and the results of ongoing human clinical trials are eagerly awaited. These dietary interventions directly influence nutrient signaling and substrate availability in the cystic kidney, while also conferring systemic metabolic benefits. The present review focuses on the importance of local and systemic metabolism in ADPKD and summarizes current evidence for dietary interventions to slow disease progression and improve quality of life., (Copyright © 2022 © The Author(s) 2021. Published by Oxford University Press on behalf of the American Society for Nutrition. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
11. Insights into Autosomal Dominant Polycystic Kidney Disease from Genetic Studies.
- Author
-
Lanktree MB, Haghighi A, di Bari I, Song X, and Pei Y
- Subjects
- Animals, Genetic Testing, Humans, Mutation, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Autosomal dominant polycystic kidney disease is the most common monogenic cause of ESKD. Genetic studies from patients and animal models have informed disease pathobiology and strongly support a "threshold model" in which cyst formation is triggered by reduced functional polycystin dosage below a critical threshold within individual tubular epithelial cells due to ( 1 ) germline and somatic PKD1 and/or PKD2 mutations, ( 2 ) mutations of genes ( e.g. , SEC63 , SEC61B , GANAB , PRKCSH , DNAJB11 , ALG8 , and ALG9 ) in the endoplasmic reticulum protein biosynthetic pathway, or ( 3 ) somatic mosaicism. Genetic testing has the potential to provide diagnostic and prognostic information in cystic kidney disease. However, mutation screening of PKD1 is challenging due to its large size and complexity, making it both costly and labor intensive. Moreover, conventional Sanger sequencing-based genetic testing is currently limited in elucidating the causes of atypical polycystic kidney disease, such as within-family disease discordance, atypical kidney imaging patterns, and discordant disease severity between total kidney volume and rate of eGFR decline. In addition, environmental factors, genetic modifiers, and somatic mosaicism also contribute to disease variability, further limiting prognostication by mutation class in individual patients. Recent innovations in next-generation sequencing are poised to transform and extend molecular diagnostics at reasonable costs. By comprehensive screening of multiple cystic disease and modifier genes, targeted gene panel, whole-exome, or whole-genome sequencing is expected to improve both diagnostic and prognostic accuracy to advance personalized medicine in autosomal dominant polycystic kidney disease., (Copyright © 2021 by the American Society of Nephrology.)
- Published
- 2021
- Full Text
- View/download PDF
12. Patients with Protein-Truncating PKD1 Mutations and Mild ADPKD.
- Author
-
Lanktree MB, Guiard E, Akbari P, Pourafkari M, Iliuta IA, Ahmed S, Haghighi A, He N, Song X, Paterson AD, Khalili K, and Pei YPC
- Subjects
- Adult, Female, Humans, Male, Middle Aged, Severity of Illness Index, Mutation, Polycystic Kidney, Autosomal Dominant genetics, TRPP Cation Channels genetics
- Abstract
Background and Objectives: Progression of autosomal dominant polycystic kidney disease (ADPKD) is highly variable. On average, protein-truncating PKD1 mutations are associated with the most severe kidney disease among all mutation classes. Here, we report that patients with protein-truncating PKD1 mutations may also have mild kidney disease, a finding not previously well recognized., Design, Setting, Participants, & Measurements: From the extended Toronto Genetic Epidemiologic Study of Polycystic Kidney Disease, 487 patients had PKD1 and PKD2 sequencing and typical ADPKD imaging patterns by magnetic resonance imaging or computed tomography. Mayo Clinic Imaging Classification on the basis of age- and height-adjusted total kidney volume was used to assess their cystic disease severity; classes 1A or 1B were used as a proxy to define mild disease. Multivariable linear regression was performed to test the effects of age, sex, and mutation classes on log-transformed height-adjusted total kidney volume and eGFR., Results: Among 174 study patients with typical imaging patterns and protein-truncating PKD1 mutations, 32 (18%) were found to have mild disease on the basis of imaging results ( i.e ., Mayo Clinic Imaging class 1A-1B), with their mutations spanning the entire gene. By multivariable analyses of age, sex, and mutation class, they displayed mild disease similar to patients with PKD2 mutations and Mayo Clinic Imaging class 1A-1B. Most of these mildly affected patients with protein-truncating PKD1 mutations reported a positive family history of ADPKD in preceding generations and displayed significant intrafamilial disease variability., Conclusions: Despite having the most severe mutation class, 18% of patients with protein-truncating PKD1 mutations had mild disease on the basis of clinical and imaging assessment., Podcast: This article contains a podcast at https://www.asn-online.org/media/podcast/CJASN/2021_02_18_CJN11100720_final.mp3., (Copyright © 2021 by the American Society of Nephrology.)
- Published
- 2021
- Full Text
- View/download PDF
13. Establishing a Core Outcome Set for Autosomal Dominant Polycystic Kidney Disease: Report of the Standardized Outcomes in Nephrology-Polycystic Kidney Disease (SONG-PKD) Consensus Workshop.
- Author
-
Cho Y, Tong A, Craig JC, Mustafa RA, Chapman A, Perrone RD, Ahn C, Fowler K, Torres V, Gansevoort RT, Ong ACM, Coolican H, Tze-Wah Kao J, Harris T, Gutman T, Shen JI, Viecelli AK, Johnson DW, Au E, El-Damanawi R, Logeman C, Ju A, Manera KE, Chonchol M, Odland D, Baron D, Pei Y, Sautenet B, Rastogi A, Sharma A, and Rangan G
- Subjects
- Activities of Daily Living, Administrative Personnel, Cardiovascular Diseases etiology, Caregivers, Delphi Technique, Disease Progression, Humans, Nephrologists, Outcome Assessment, Health Care, Pain etiology, Polycystic Kidney, Autosomal Dominant complications, Polycystic Kidney, Autosomal Dominant therapy, Renal Insufficiency etiology, Stakeholder Participation, Cardiovascular Diseases physiopathology, Mortality, Pain physiopathology, Polycystic Kidney, Autosomal Dominant physiopathology, Renal Insufficiency physiopathology
- Abstract
The omission of outcomes that are of relevance to patients, clinicians, and regulators across trials in autosomal dominant polycystic kidney disease (ADPKD) limits shared decision making. The Standardized Outcomes in Nephrology-Polycystic Kidney Disease (SONG-PKD) Initiative convened an international consensus workshop on October 25, 2018, to discuss the identification and implementation of a potential core outcome set for all ADPKD trials. This article summarizes the discussion from the workshops and the SONG-PKD core outcome set. Key stakeholders including 11 patients/caregivers and 47 health professionals (nephrologists, policy makers, industry, and researchers) attended the workshop. Four themes emerged: "Relevance of trajectory and impact of kidney function" included concerns about a patient's prognosis and uncertainty of when they may need to commence kidney replacement therapy and the lack of an early prognostic marker to inform long-term decisions; "Discerning and defining pain specific to ADPKD" highlighted the challenges in determining the origin of pain, adapting to the chronicity and repeated episodes of pain, the need to place emphasis on pain management, and to have a validated measure for pain; "Highlighting ADPKD consequences" encompassed cyst-related complications and reflected patient's knowledge because of family history and the hereditary nature of ADPKD; and "Risk for life-threatening but rare consequences" such as cerebral aneurysm meant considering both frequency and severity of the outcome. Kidney function, mortality, cardiovascular disease, and pain were established as the core outcomes for ADPKD., (Copyright © 2020 National Kidney Foundation, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
14. 'A sword of Damocles': patient and caregiver beliefs, attitudes and perspectives on presymptomatic testing for autosomal dominant polycystic kidney disease: a focus group study.
- Author
-
Logeman C, Cho Y, Sautenet B, Rangan GK, Gutman T, Craig J, Ong A, Chapman A, Ahn C, Coolican H, Tze-Wah Kao J, Gansevoort RT, Perrone R, Harris T, Torres V, Fowler K, Pei Y, Kerr P, Ryan J, Johnson D, Viecelli A, Geneste C, Kim H, Kim Y, Howell M, Ju A, Manera KE, Teixeira-Pinto A, Parasivam G, and Tong A
- Subjects
- Adolescent, Adult, Attitude, Australia, Focus Groups, France, Humans, Quality of Life, Republic of Korea, Caregivers, Polycystic Kidney, Autosomal Dominant diagnosis
- Abstract
Background and Objectives: Presymptomatic testing is available for early diagnosis of hereditary autosomal dominant polycystic kidney disease (ADPKD). However, the complex ethical and psychosocial implications can make decision-making challenging and require an understanding of patients' values, goals and priorities. This study aims to describe patient and caregiver beliefs and expectations regarding presymptomatic testing for ADPKD., Design, Setting and Participants: 154 participants (120 patients and 34 caregivers) aged 18 years and over from eight centres in Australia, France and Korea participated in 17 focus groups. Transcripts were analysed thematically., Results: We identified five themes: avoiding financial disadvantage (insecurity in the inability to obtain life insurance, limited work opportunities, financial burden); futility in uncertainty (erratic and diverse manifestations of disease limiting utility, taking preventive actions in vain, daunted by perplexity of results, unaware of risk of inheriting ADPKD); lacking autonomy and support in decisions (overwhelmed by ambiguous information, medicalising family planning, family pressures); seizing control of well-being (gaining confidence in early detection, allowing preparation for the future, reassurance in family resilience); and anticipating impact on quality of life (reassured by lack of symptoms, judging value of life with ADPKD)., Conclusions: For patients with ADPKD, presymptomatic testing provides an opportunity to take ownership of their health through family planning and preventive measures. However, these decisions can be wrought with tensions and uncertainty about prognostic implications, and the psychosocial and financial burden of testing. Healthcare professionals should focus on genetic counselling, mental health and providing education to patients' families to support informed decision-making. Policymakers should consider the cost burden and risk of discrimination when informing government policies. Finally, patients are recommended to focus on self-care from an early age., Competing Interests: Competing interests: GKR declares he is site investigator of clinical trials sponsored by Sanofi, Otsuka and Reata, principal investigator of the PREVENT-ADPKD clinical trial (funded in part by the NHMRC, Danone Nutricia—manufacturer of bottled water—PKD Australia, Westmead Medical Research Foundation) and Chair, Scientific Advisory Board, PKD Australia. RP declares he is site investigator of clinical trials sponsored by Sanofi, Kadmon, Reata, Otsuka and the US Department of Defense (TAME PKD), and section editor for Cystic Kidney Disease, UpToDate., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
15. Core Outcome Domains for Trials in Autosomal Dominant Polycystic Kidney Disease: An International Delphi Survey.
- Author
-
Cho Y, Rangan G, Logeman C, Ryu H, Sautenet B, Perrone RD, Nadeau-Fredette AC, Mustafa RA, Htay H, Chonchol M, Harris T, Gutman T, Craig JC, Ong ACM, Chapman A, Ahn C, Coolican H, Kao JT, Gansevoort RT, Torres V, Pei Y, Johnson DW, Viecelli AK, Teixeira-Pinto A, Howell M, Ju A, Manera KE, and Tong A
- Subjects
- Adolescent, Adult, Africa epidemiology, Aged, Asia epidemiology, Caregivers psychology, Child, Consensus, Delphi Technique, Female, Health Personnel psychology, Humans, Intracranial Aneurysm etiology, Kidney Failure, Chronic epidemiology, Kidney Failure, Chronic etiology, Male, Middle Aged, Pain etiology, Patient Reported Outcome Measures, Patients psychology, Polycystic Kidney, Autosomal Dominant complications, Polycystic Kidney, Autosomal Dominant epidemiology, Polycystic Kidney, Autosomal Dominant psychology, Qualitative Research, Quality of Life, Self Concept, Socioeconomic Factors, Stress, Psychological, Young Adult, Polycystic Kidney, Autosomal Dominant therapy
- Abstract
Rationale & Objective: Outcomes reported in trials involving patients with autosomal dominant polycystic kidney disease (ADPKD) are heterogeneous and rarely include patient-reported outcomes. We aimed to identify critically important consensus-based core outcome domains to be reported in trials in ADPKD., Study Design: An international 2-round online Delphi survey was conducted in English, French, and Korean languages., Setting & Participants: Patients/caregivers and health professionals completed a 9-point Likert scale (7-9 indicating critical importance) and a Best-Worst Scale., Analytical Approach: The absolute and relative importance of outcomes were assessed. Comments were analyzed thematically., Results: 1,014 participants (603 [60%] patients/caregivers, 411 [40%] health professionals) from 56 countries completed round 1, and 713 (70%) completed round 2. The prioritized outcomes were kidney function (importance score, 8.6), end-stage kidney disease (8.6), death (7.9), blood pressure (7.9), kidney cyst size/growth (7.8), and cerebral aneurysm (7.7). Kidney cyst-related pain was the highest rated patient-reported outcome by both stakeholder groups. Seven themes explained the prioritization of outcomes: protecting life and health, directly encountering life-threatening and debilitating consequences, specificity to ADPKD, optimizing and extending quality of life, hidden suffering, destroying self-confidence, and lost opportunities., Limitations: Study design precluded involvement from those without access to internet or limited computer literacy., Conclusions: Kidney function, end-stage kidney disease, and death were the most important outcomes to patients, caregivers, and health professionals. Kidney cyst-related pain was the highest rated patient-reported outcome. Consistent reporting of these top prioritized outcomes may strengthen the value of trials in ADPKD for decision making., (Copyright © 2020 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
16. Targeting AMP-activated protein kinase (AMPK) for treatment of autosomal dominant polycystic kidney disease.
- Author
-
Song X, Tsakiridis E, Steinberg GR, and Pei Y
- Subjects
- AMP-Activated Protein Kinase Kinases, Animals, Cell Proliferation, Energy Metabolism, Humans, Signal Transduction, Mechanistic Target of Rapamycin Complex 1 antagonists & inhibitors, Polycystic Kidney, Autosomal Dominant drug therapy, Polycystic Kidney, Autosomal Dominant metabolism, Protein Kinases metabolism
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenetic kidney disease worldwide and an important cause of chronic kidney disease. Multiple experimental studies have highlighted the role of increased mammalian target of rapamycin complex 1 (mTORC1) and reduced AMP-activated protein kinase (AMPK) signaling in modulating cyst growth in ADPKD. Notably, mTORC1 and AMPK are two diametrically opposing sensors of energy metabolism which regulate cell growth and proliferation. Although pharmacological mTORC1 inhibition was highly effective in experimental studies of ADPKD, clinical trials of mTORC1 inhibitors showed a lack of efficacy with low-dose treatment and poor tolerability with high-dose treatment. Therapeutic AMPK activation has been shown to attenuate cystic kidney disease severity in Pkd1 mutant animal models by improving mitochondrial biogenesis and reducing tissue inflammation. This review summarizes the current knowledge on the function of AMPK as a regulator of cellular energy metabolism and how AMPK activation by pharmacological and non-pharmacological means can potentially be exploited to treat ADPKD in the clinical settings., Competing Interests: Declaration of Competing Interest Y.P. received compensation for participating in advisory and consultancy boards with Otsuka pharmaceuticals. G.R.S. has received honoraria/consulting fees from Esperion Therapeutics and Poxel, research funding from Esperion Therapeutics and Espervita Therapeutics and is a shareholder of Espervita Therapeutics., (Copyright © 2020 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
17. Range and Variability of Outcomes Reported in Randomized Trials Conducted in Patients With Polycystic Kidney Disease: A Systematic Review.
- Author
-
Sautenet B, Cho Y, Gutman T, Rangan G, Ong A, Chapman AB, Ahn C, Coolican H, Tze-Wah Kao J, Fowler K, Gansevoort RT, Geneste C, Perrone RD, Harris T, Torres VE, Pei Y, Craig JC, and Tong A
- Subjects
- Blood Pressure, Cardiovascular Diseases epidemiology, Humans, Infections epidemiology, Kidney Function Tests, Organ Size, Pain epidemiology, Patient Reported Outcome Measures, Polycystic Kidney, Autosomal Dominant metabolism, Polycystic Kidney, Autosomal Dominant physiopathology, Renal Insufficiency epidemiology, Renal Insufficiency therapy, Clinical Trials as Topic, Outcome Assessment, Health Care, Polycystic Kidney, Autosomal Dominant therapy
- Abstract
Rationale & Objective: Trials in autosomal dominant polycystic kidney disease (ADPKD) have increased, but their impact on decision making has been limited. Because heterogeneity in reported outcomes may be responsible, we assessed their range and variability in ADPKD trials., Study Design: Systematic review., Setting & Study Population: Adult participants in clinical trials in ADPKD., Selection Criteria for Studies: We included trials that studied adults and were published in English. For trials that enrolled patients without ADPKD, only those enrolling ≥50% of participants with ADPKD were included., Data Extraction: We extracted information on all discrete outcome measures, grouped them into 97 domains, and classified them into clinical, surrogate, and patient-reported categories. For each category, we choose the 3 most frequently reported domains and performed a detailed analysis of outcome measures., Analytical Approach: Frequencies and characteristics of outcome measures were described., Results: Among 68 trials, 1,413 different outcome measures were reported. 97 domains were identified; 41 (42%) were surrogate, 30 (31%) were clinical, and 26 (27%) were patient reported. The 3 most frequently reported domains were in the surrogate category: kidney function (54; 79% of trials; using 46 measures), kidney and cyst volumes (43; 63% of trials; 52 measures), and blood pressure (27; 40% of trials, 30 measures); in the clinical category: infection (10; 15%; 21 measures), cardiovascular events (9; 13%; 6 measures), and kidney failure requiring kidney replacement therapy (8; 12%; 5 measures); and in the patient-reported category: pain related to ADPKD (16; 24%; 26 measures), pain for other reasons (11; 16%; 11 measures), and diarrhea/constipation/gas (10; 15%; 9 measures)., Limitations: Outcome measures were assessed for only the top 3 domains in each category., Conclusions: The outcomes in ADPKD trials are broad in scope and highly variable. Surrogate outcomes were most frequently reported. Patient-reported outcomes were uncommon. A consensus-based set of core outcomes meaningful to patients and clinicians is needed for future ADPKD trials., (Copyright © 2019 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
18. Global microRNA profiling in human urinary exosomes reveals novel disease biomarkers and cellular pathways for autosomal dominant polycystic kidney disease.
- Author
-
Magayr TA, Song X, Streets AJ, Vergoz L, Chang L, Valluru MK, Yap HL, Lannoy M, Haghighi A, Simms RJ, Tam FWK, Pei Y, and Ong ACM
- Subjects
- Animals, Biomarkers, Gene Expression Profiling, Humans, Kidney, Mice, Exosomes genetics, MicroRNAs genetics, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
MicroRNAs (miRNAs) play an important role in regulating gene expression in health and disease but their role in modifying disease expression in Autosomal Dominant Polycystic Kidney Disease (ADPKD) remains uncertain. Here, we profiled human urinary exosome miRNA by global small RNA-sequencing in an initial discovery cohort of seven patients with ADPKD with early disease (eGFR over 60ml/min/1.73m
2 ), nine with late disease (eGFR under 60ml/min/1.73m2 ), and compared their differential expression with six age and sex matched healthy controls. Two kidney-enriched candidate miRNA families were identified (miR-192/miR-194-2 and miR-30) and selected for confirmatory testing in a 60 patient validation cohort by quantitative polymerase chain reaction. We confirmed that miR-192-5p, miR-194-5p, miR-30a-5p, miR-30d-5p and miR-30e-5p were significantly downregulated in patient urine exosomes, in murine Pkd1 cystic kidneys and in human PKD1 cystic kidney tissue. All five miRNAs showed significant correlations with baseline eGFR and ultrasound-determined mean kidney length and improved the diagnostic performance (area under the curve) of mean kidney length for the rate of disease progression. Finally, inverse correlations of these two miRNA families with increased expression in their predicted target genes in patient PKD1 cystic tissue identified dysregulated pathways and transcriptional networks including novel interactions between miR-194-5p and two potentially relevant candidate genes, PIK3R1 and ANO1. Thus, our results identify a subset of urinary exosomal miRNAs that could serve as novel biomarkers of disease progression and suggest new therapeutic targets in ADPKD., (Copyright © 2020 International Society of Nephrology. All rights reserved.)- Published
- 2020
- Full Text
- View/download PDF
19. Next-generation sequencing for detection of somatic mosaicism in autosomal dominant polycystic kidney disease.
- Author
-
Devuyst O and Pei Y
- Subjects
- High-Throughput Nucleotide Sequencing, Humans, Mosaicism, Mutation, TRPP Cation Channels genetics, Polycystic Kidney, Autosomal Dominant
- Abstract
Mosaicism is defined as the presence of 2 genetically different populations of cells in a single organism, resulting from a mutation during early embryogenesis. Hopp et al. characterized mosaicism in 20 unresolved ADPKD families, using next-generation sequencing techniques with DNA isolated from blood cells. Mosaicism may be involved in 1% of ADPKD families, and it may explain some atypical disease phenotypes., (Copyright © 2020 International Society of Nephrology. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
20. Identifying patient-important outcomes in polycystic kidney disease: An international nominal group technique study.
- Author
-
Cho Y, Sautenet B, Gutman T, Rangan G, Craig JC, Ong AC, Chapman A, Ahn C, Coolican H, Kao JT, Gansevoort R, Perrone RD, Harris T, Torres V, Pei Y, Kerr PG, Ryan J, Johnson DW, Viecelli AK, Geneste C, Kim H, Kim Y, Oh YK, Teixeira-Pinto A, Logeman C, Howell M, Ju A, Manera KE, and Tong A
- Subjects
- Attitude to Health, Australia, Caregivers psychology, Disease Progression, Evaluation Studies as Topic, Female, France, Humans, Kidney Function Tests psychology, Male, Middle Aged, Progression-Free Survival, Republic of Korea, Cost of Illness, Kidney Failure, Chronic etiology, Kidney Failure, Chronic mortality, Life Style, Patient Outcome Assessment, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant epidemiology, Polycystic Kidney, Autosomal Dominant physiopathology, Polycystic Kidney, Autosomal Dominant psychology, Quality of Life
- Abstract
Aim: Patients with autosomal dominant polycystic kidney disease (ADPKD) are at increased risk of premature mortality, morbidities and complications, which severely impair quality of life. However, patient-centered outcomes are not consistently reported in trials in ADPKD, which can limit shared decision-making. We aimed to identify outcomes important to patients and caregivers and the reasons for their priorities., Methods: Nominal group technique was adopted involving patients with ADPKD and caregivers who were purposively selected from eight centres across Australia, France and the Republic of Korea. Participants identified, ranked and discussed outcomes for trials in ADPKD. We calculated an importance score (0-1) for each outcome and conducted thematic analyses., Results: Across 17 groups, 154 participants (121 patients, 33 caregivers) aged 19 to 78 (mean 54.5 years) identified 55 outcomes. The 10 highest ranked outcomes were: kidney function (importance score 0.36), end-stage kidney disease (0.32), survival (0.21), cyst size/growth (0.20), cyst pain/bleeding (0.18), blood pressure (0.17), ability to work (0.16), cerebral aneurysm/stroke (0.14), mobility/physical function (0.12), and fatigue (0.12). Three themes were identified: threatening semblance of normality, inability to control and making sense of diverse risks., Conclusion: For patients with ADPKD and their caregivers, kidney function, delayed progression to end-stage kidney disease and survival were the highest priorities, and were focused on achieving normality, and maintaining control over health and lifestyle. Implementing these patient-important outcomes may improve the meaning and relevance of trials to inform clinical care in ADPKD., (© 2019 Asian Pacific Society of Nephrology.)
- Published
- 2019
- Full Text
- View/download PDF
21. Evolving role of genetic testing for the clinical management of autosomal dominant polycystic kidney disease.
- Author
-
Lanktree MB, Iliuta IA, Haghighi A, Song X, and Pei Y
- Subjects
- Humans, Mutation, Polycystic Kidney, Autosomal Dominant genetics, Prognosis, Biomarkers analysis, Genetic Testing methods, Polycystic Kidney, Autosomal Dominant diagnosis
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is caused primarily by mutations of two genes, PKD1 and PKD2. In the presence of a positive family history of ADPKD, genetic testing is currently seldom indicated as the diagnosis is mostly based on imaging studies using well-established criteria. Moreover, PKD1 mutation screening is technically challenging due to its large size, complexity (i.e. presence of six pseudogenes with high levels of DNA sequence similarity) and extensive allelic heterogeneity. Despite these limitations, recent studies have delineated a strong genotype-phenotype correlation in ADPKD and begun to unravel the role of genetics underlying cases with atypical phenotypes. Furthermore, adaptation of next-generation sequencing (NGS) to clinical PKD genetic testing will provide a high-throughput, accurate and comprehensive screen of multiple cystic disease and modifier genes at a reduced cost. In this review, we discuss the evolving indications of genetic testing in ADPKD and how NGS-based screening promises to yield clinically important prognostic information for both typical as well as unusual genetic (e.g. allelic or genic interactions, somatic mosaicism, cystic kidney disease modifiers) cases to advance personalized medicine in the era of novel therapeutics for ADPKD., (© The Author(s) 2018. Published by Oxford University Press on behalf of ERA-EDTA. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
22. Prognostic Performance of Kidney Volume Measurement for Polycystic Kidney Disease: A Comparative Study of Ellipsoid vs. Manual Segmentation.
- Author
-
Shi B, Akbari P, Pourafkari M, Iliuta IA, Guiard E, Quist CF, Song X, Hillier D, Khalili K, and Pei Y
- Subjects
- Adult, Cohort Studies, Cross-Sectional Studies, Disease Progression, Female, Humans, Image Processing, Computer-Assisted methods, Kidney pathology, Male, Middle Aged, Organ Size, Polycystic Kidney, Autosomal Dominant pathology, Prognosis, Kidney diagnostic imaging, Magnetic Resonance Imaging methods, Polycystic Kidney, Autosomal Dominant diagnostic imaging
- Abstract
Total kidney volume (TKV) is a validated prognostic biomarker for risk assessment in autosomal dominant polycystic kidney disease (ADPKD). TKV by manual segmentation (MS) is the "gold standard" but is time-consuming and requires expertise. The purpose of this study was to compare TKV-based prognostic performance by ellipsoid (EL) vs. MS in a large cohort of patients. Cross-sectional study of 308 patients seen at a tertiary referral center; all had a standardized MRI with typical imaging of ADPKD. An experienced radiologist blinded to patient clinical results performed all TKV measurements by EL and MS. We assessed the agreement of TKV measurements by intraclass correlation(ICC) and Bland-Altman plot and also how the disagreement of the two methods impact the prognostic performance of the Mayo Clinic Imaging Classification (MCIC). We found a high ICC of TKV measurements (0.991, p < 0.001) between EL vs. MS; however, 5.5% of the cases displayed disagreement of TKV measurements >20%. We also found a high degree of agreement of the individual MCIC risk classes (i.e. 1A to 1E) with a Cohen's weighted-kappa of 0.89; but 42 cases (13.6%) were misclassified by EL with no misclassification spanning more than one risk class. The sensitivity and specificity of EL in distinguishing low-risk (1A-B) from high-risk (1C-E) MCIC prognostic grouping were 96.6% and 96.1%, respectively. Overall, we found an excellent agreement of TKV-based risk assessment between EL and MS. However, caution is warranted for patients with MCIC 1B and 1C, as misclassification can have therapeutic consequence.
- Published
- 2019
- Full Text
- View/download PDF
23. Prevalence Estimates of Polycystic Kidney and Liver Disease by Population Sequencing.
- Author
-
Lanktree MB, Haghighi A, Guiard E, Iliuta IA, Song X, Harris PC, Paterson AD, and Pei Y
- Subjects
- Computational Biology, Databases, Genetic, Female, Genes, Modifier, Genetic Variation, Humans, Male, Mutation, Prevalence, Risk Factors, TRPP Cation Channels genetics, Exome Sequencing, Whole Genome Sequencing, Cysts epidemiology, Cysts genetics, Liver Diseases epidemiology, Liver Diseases genetics, Polycystic Kidney, Autosomal Dominant epidemiology, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Background: Estimating the prevalence of autosomal dominant polycystic kidney disease (ADPKD) is challenging because of age-dependent penetrance and incomplete clinical ascertainment. Early studies estimated the lifetime risk of ADPKD to be about one per 1000 in the general population, whereas recent epidemiologic studies report a point prevalence of three to five cases per 10,000 in the general population., Methods: To measure the frequency of high-confidence mutations presumed to be causative in ADPKD and autosomal dominant polycystic liver disease (ADPLD) and estimate lifetime ADPKD prevalence, we used two large, population sequencing databases, gnomAD (15,496 whole-genome sequences; 123,136 exome sequences) and BRAVO (62,784 whole-genome sequences). We used stringent criteria for defining rare variants in genes involved in ADPKD ( PKD1 , PKD2 ), ADPLD ( PRKCSH , SEC63 , GANAB , ALG8 , SEC61B , LRP5 ), and potential cystic disease modifiers; evaluated variants for quality and annotation; compared variants with data from an ADPKD mutation database; and used bioinformatic tools to predict pathogenicity., Results: Identification of high-confidence pathogenic mutations in whole-genome sequencing provided a lower boundary for lifetime ADPKD prevalence of 9.3 cases per 10,000 sequenced. Estimates from whole-genome and exome data were similar. Truncating mutations in ADPLD genes and genes of potential relevance as cyst modifiers were found in 20.2 cases and 103.9 cases per 10,000 sequenced, respectively., Conclusions: Population whole-genome sequencing suggests a higher than expected prevalence of ADPKD-associated mutations. Loss-of-function mutations in ADPLD genes are also more common than expected, suggesting the possibility of unrecognized cases and incomplete penetrance. Substantial rare variation exists in genes with potential for phenotype modification in ADPKD., (Copyright © 2018 by the American Society of Nephrology.)
- Published
- 2018
- Full Text
- View/download PDF
24. Monoallelic Mutations to DNAJB11 Cause Atypical Autosomal-Dominant Polycystic Kidney Disease.
- Author
-
Cornec-Le Gall E, Olson RJ, Besse W, Heyer CM, Gainullin VG, Smith JM, Audrézet MP, Hopp K, Porath B, Shi B, Baheti S, Senum SR, Arroyo J, Madsen CD, Férec C, Joly D, Jouret F, Fikri-Benbrahim O, Charasse C, Coulibaly JM, Yu AS, Khalili K, Pei Y, Somlo S, Le Meur Y, Torres VE, and Harris PC
- Subjects
- Adult, Aged, Aged, 80 and over, Amino Acid Sequence, Base Sequence, Epithelial Cells metabolism, Family, Female, HSP40 Heat-Shock Proteins chemistry, Humans, Loop of Henle pathology, Male, Middle Aged, Pedigree, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Polycystic Kidney, Autosomal Dominant pathology, TRPP Cation Channels genetics, Uromodulin metabolism, Exome Sequencing, Young Adult, Alleles, HSP40 Heat-Shock Proteins genetics, Mutation genetics, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is characterized by the progressive development of kidney cysts, often resulting in end-stage renal disease (ESRD). This disorder is genetically heterogeneous with ∼7% of families genetically unresolved. We performed whole-exome sequencing (WES) in two multiplex ADPKD-like pedigrees, and we analyzed a further 591 genetically unresolved, phenotypically similar families by targeted next-generation sequencing of 65 candidate genes. WES identified a DNAJB11 missense variant (p.Pro54Arg) in two family members presenting with non-enlarged polycystic kidneys and a frameshifting change (c.166_167insTT) in a second family with small renal and liver cysts. DNAJB11 is a co-factor of BiP, a key chaperone in the endoplasmic reticulum controlling folding, trafficking, and degradation of secreted and membrane proteins. Five additional multigenerational families carrying DNAJB11 mutations were identified by the targeted analysis. The clinical phenotype was consistent in the 23 affected members, with non-enlarged cystic kidneys that often evolved to kidney atrophy; 7 subjects reached ESRD from 59 to 89 years. The lack of kidney enlargement, histologically evident interstitial fibrosis in non-cystic parenchyma, and recurring episodes of gout (one family) suggested partial phenotypic overlap with autosomal-dominant tubulointerstitial diseases (ADTKD). Characterization of DNAJB11-null cells and kidney samples from affected individuals revealed a pathogenesis associated with maturation and trafficking defects involving the ADPKD protein, PC1, and ADTKD proteins, such as UMOD. DNAJB11-associated disease is a phenotypic hybrid of ADPKD and ADTKD, characterized by normal-sized cystic kidneys and progressive interstitial fibrosis resulting in late-onset ESRD., (Copyright © 2018 American Society of Human Genetics. Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
25. Standardised Outcomes in Nephrology-Polycystic Kidney Disease (SONG-PKD): study protocol for establishing a core outcome set in polycystic kidney disease.
- Author
-
Cho Y, Sautenet B, Rangan G, Craig JC, Ong ACM, Chapman A, Ahn C, Chen D, Coolican H, Kao JT, Gansevoort R, Perrone R, Harris T, Torres V, Pei Y, Kerr PG, Ryan J, Gutman T, Howell M, Ju A, Manera KE, Teixeira-Pinto A, Hamiwka LA, and Tong A
- Subjects
- Consensus, Delphi Technique, Humans, Nephrology methods, Polycystic Kidney, Autosomal Dominant diagnosis, Stakeholder Participation, Systematic Reviews as Topic, Treatment Outcome, Endpoint Determination standards, Nephrology standards, Patient Outcome Assessment, Polycystic Kidney, Autosomal Dominant therapy, Research Design standards
- Abstract
Background: Autosomal dominant polycystic kidney disease (ADPKD) is the most common potentially life threatening inherited kidney disease and is responsible for 5-10% of cases of end-stage kidney disease (ESKD). Cystic kidneys may enlarge up to 20 times the weight of a normal kidney due to the growth of renal cysts, and patients with ADPKD have an increased risk of morbidity, premature mortality, and other life-time complications including renal and hepatic cyst and urinary tract infection, intracranial aneurysm, diverticulosis, and kidney pain which impair quality of life. Despite some therapeutic advances and the growing number of clinical trials in ADPKD, the outcomes that are relevant to patients and clinicians, such as symptoms and quality of life, are infrequently and inconsistently reported. This potentially limits the contribution of trials to inform evidence-based decision-making. The Standardised Outcomes in Nephrology-Polycystic Kidney Disease (SONG-PKD) project aims to establish a consensus-based set of core outcomes for trials in PKD (with an initial focus on ADPKD but inclusive of all stages) that patients and health professionals identify as critically important., Methods: The five phases of SONG-PKD are: a systematic review to identify outcomes that have been reported in existing PKD trials; focus groups with nominal group technique with patients and caregivers to identify, rank, and describe reasons for their choices; qualitative stakeholder interviews with health professionals to elicit individual values and perspectives on outcomes for trials involving patients with PKD; an international three-round Delphi survey with all stakeholder groups (including patients, caregivers, healthcare providers, policy makers, researchers, and industry) to gain consensus on critically important core outcome domains; and a consensus workshop to review and establish a set of core outcome domains and measures for trials in PKD., Discussion: The SONG-PKD core outcome set is aimed at improving the consistency and completeness of outcome reporting across ADPKD trials, leading to improvements in the reliability and relevance of trial-based evidence to inform decisions about treatment and ultimately improve the care and outcomes for people with ADPKD.
- Published
- 2017
- Full Text
- View/download PDF
26. Bosutinib versus Placebo for Autosomal Dominant Polycystic Kidney Disease.
- Author
-
Tesar V, Ciechanowski K, Pei Y, Barash I, Shannon M, Li R, Williams JH, Levisetti M, Arkin S, and Serra A
- Subjects
- Adolescent, Adult, Aniline Compounds adverse effects, Double-Blind Method, Female, Humans, Male, Middle Aged, Nitriles adverse effects, Quinolines adverse effects, Young Adult, Aniline Compounds therapeutic use, Nitriles therapeutic use, Polycystic Kidney, Autosomal Dominant drug therapy, Quinolines therapeutic use
- Abstract
Overactivation of Src has been linked to the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD). This phase 2, multisite study assessed the efficacy and safety of bosutinib, an oral dual Src/Bcr-Abl tyrosine kinase inhibitor, in patients with ADPKD. Patients with ADPKD, eGFR≥60 ml/min per 1.73 m
2 , and total kidney volume ≥750 ml were randomized 1:1:1 to bosutinib 200 mg/d, bosutinib 400 mg/d, or placebo for ≤24 months. The primary endpoint was annualized rate of kidney enlargement in patients treated for ≥2 weeks who had at least one postbaseline magnetic resonance imaging scan that was preceded by a 30-day washout (modified intent-to-treat population). Of 172 enrolled patients, 169 received at least one study dose. Per protocol amendment, doses for 24 patients who initially received bosutinib at 400 mg/d were later reduced to 200 mg/d. The annual rate of kidney enlargement was reduced by 66% for bosutinib 200 mg/d versus placebo (1.63% versus 4.74%, respectively; P =0.01) and by 82% for pooled bosutinib versus placebo (0.84% versus 4.74%, respectively; P <0.001). Over the treatment period, patients receiving placebo or bosutinib had similar annualized eGFR decline. Gastrointestinal and liver-related adverse events were the most frequent toxicities. In conclusion, compared with placebo, bosutinib at 200 mg/d reduced kidney growth in patients with ADPKD. The overall gastrointestinal and liver toxicity profile was consistent with the profile in prior studies of bosutinib; no new toxicities were identified. (ClinicalTrials.gov: NCT01233869)., (Copyright © 2017 by the American Society of Nephrology.)- Published
- 2017
- Full Text
- View/download PDF
27. Molecular diagnosis of autosomal dominant polycystic kidney disease.
- Author
-
Song X, Haghighi A, Iliuta IA, and Pei Y
- Subjects
- Diagnosis, Differential, Genetic Markers, Genomics methods, High-Throughput Nucleotide Sequencing, Humans, Mutation, Polycystic Kidney, Autosomal Dominant metabolism, Polycystic Kidney, Autosomal Dominant mortality, Prognosis, TRPP Cation Channels genetics, TRPP Cation Channels metabolism, Genetic Association Studies, Genetic Testing methods, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics
- 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).
- Published
- 2017
- Full Text
- View/download PDF
28. Polycystic Kidney Disease without an Apparent Family History.
- Author
-
Iliuta IA, Kalatharan V, Wang K, Cornec-Le Gall E, Conklin J, Pourafkari M, Ting R, Chen C, Borgo AC, He N, Song X, Heyer CM, Senum SR, Hwang YH, Paterson AD, Harris PC, Khalili K, and Pei Y
- Subjects
- Adult, Aged, Aged, 80 and over, DNA Mutational Analysis, Female, Germ-Line Mutation, Humans, Male, Medical History Taking, Middle Aged, Mosaicism, Parents, Pedigree, Polycystic Kidney, Autosomal Dominant physiopathology, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Polycystic Kidney, Autosomal Dominant genetics, TRPP Cation Channels genetics
- Abstract
The absence of a positive family history (PFH) in 10%-25% of patients poses a diagnostic challenge for autosomal dominant polycystic kidney disease (ADPKD). In the Toronto Genetic Epidemiology Study of Polycystic Kidney Disease, 210 affected probands underwent renal function testing, abdominal imaging, and comprehensive PKD1 and PKD2 mutation screening. From this cohort, we reviewed all patients with and without an apparent family history, examined their parental medical records, and performed renal imaging in all available parents of unknown disease status. Subsequent reclassification of 209 analyzed patients revealed 72.2% (151 of 209) with a PFH, 15.3% (32 of 209) with de novo disease, 10.5% (22 of 209) with an indeterminate family history, and 1.9% (four of 209) with PFH in retrospect. Among the patients with de novo cases, we found two families with germline mosaicism and one family with somatic mosaicism. Additionally, analysis of renal imaging revealed that 16.3% (34 of 209) of patients displayed atypical PKD, most of which followed one of three patterns: asymmetric or focal PKD with PFH and an identified PKD1 or PKD2 mutation (15 of 34), asymmetric and de novo PKD with proven or suspected somatic mosaicism (seven of 34), or focal PKD without any identifiable PKD1 or PKD2 mutation (eight of 34). In conclusion, PKD without an apparent family history may be due to de novo disease, missing parental medical records, germline or somatic mosaicism, or mild disease from hypomorphic PKD1 and PKD2 mutations. Furthermore, mutations of a newly identified gene for ADPKD, GANAB , and somatic mosaicism need to be considered in the mutation-negative patients with focal disease., (Copyright © 2017 by the American Society of Nephrology.)
- Published
- 2017
- Full Text
- View/download PDF
29. Methodological issues in clinical trials of polycystic kidney disease: a focused review.
- Author
-
Iliuta IA, Kitchlu A, and Pei Y
- Subjects
- Adolescent, Adult, Child, Female, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Mutation, Patient Compliance, Patient Dropouts, Patient Selection, Phenotype, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant physiopathology, TRPP Cation Channels genetics, Treatment Outcome, Young Adult, Polycystic Kidney, Autosomal Dominant therapy, Randomized Controlled Trials as Topic methods, Research Design
- Abstract
The field of therapeutics in autosomal dominant polycystic kidney disease (ADPKD) has seen a significant expansion recently, as major clinical trials have provided promising evidence in favor of new disease-modifying drugs. Though these trials are encouraging, limitations are noticeable in the form of methodological issues that restrict the interpretation of results. In this review, we discuss the methodological pitfalls of high-profile clinical interventional trials for ADPKD which have been published since 2009. Issues in study design, patient selection and follow-up, analyses and reporting of results are presented. From this review, we highlight a number of suggestions for future improvement including designs to enrich a more homogeneous patient population (i.e. based on their age-adjusted total kidney volume and/or underlying mutation class) at high-risk for disease progression, appropriate study duration and patient sample size that are matched to the disease severity of the study patients, and the use of baseline characteristics (i.e. renal function, TKV, and the proportion of PKD1 and PKD2 patients) of the analyzed patients as a quality control measure to assess any potential imbalance in randomization. Furthermore, the recognition that TKV change is not a linear trait is important in both the study design and interpretation. Implementing these lessons learned from the published trials will greatly enhance the robustness and validity of future clinical trials in ADPKD.
- Published
- 2017
- Full Text
- View/download PDF
30. Refining Genotype-Phenotype Correlation in Autosomal Dominant Polycystic Kidney Disease.
- Author
-
Hwang YH, Conklin J, Chan W, Roslin NM, Liu J, He N, Wang K, Sundsbak JL, Heyer CM, Haider M, Paterson AD, Harris PC, and Pei Y
- Subjects
- Adult, Female, Humans, Kidney physiopathology, Male, Pedigree, Polycystic Kidney, Autosomal Dominant physiopathology, Prospective Studies, Genetic Association Studies methods, Mutation, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Renal disease variability in autosomal dominant polycystic kidney disease (ADPKD) is strongly influenced by the gene locus (PKD1 versus PKD2). Recent studies identified nontruncating PKD1 mutations in approximately 30% of patients who underwent comprehensive mutation screening, but the clinical significance of these mutations is not well defined. We examined the genotype-renal function correlation in a prospective cohort of 220 unrelated ADPKD families ascertained through probands with serum creatinine ≤1.4 mg/dl at recruitment. We screened these families for PKD1 and PKD2 mutations and reviewed the clinical outcomes of the probands and affected family members. Height-adjusted total kidney volume (htTKV) was obtained in 161 affected subjects. Multivariate Cox proportional hazard modeling for renal and patient survival was performed in 707 affected probands and family members. Overall, we identified pathogenic mutations in 84.5% of our families, in which the prevalence of PKD1 truncating, PKD1 in-frame insertion/deletion, PKD1 nontruncating, and PKD2 mutations was 38.3%, 4.3%, 27.1%, and 30.3%, respectively. Compared with patients with PKD1 truncating mutations, patients with PKD1 in-frame insertion/deletion, PKD1 nontruncating, or PKD2 mutations have smaller htTKV and reduced risks (hazard ratio [95% confidence interval]) of ESRD (0.35 [0.14 to 0.91], 0.10 [0.05 to 0.18], and 0.03 [0.01 to 0.05], respectively) and death (0.31 [0.11 to 0.87], 0.20 [0.11 to 0.38], and 0.18 [0.11 to 0.31], respectively). Refined genotype-renal disease correlation coupled with targeted next generation sequencing of PKD1 and PKD2 may provide useful clinical prognostication for ADPKD., (Copyright © 2016 by the American Society of Nephrology.)
- Published
- 2016
- Full Text
- View/download PDF
31. International Multi-Specialty Delphi Survey: Identification of Diagnostic Criteria for Hepatic and Renal Cyst Infection.
- Author
-
Lantinga MA, Darding AJ, de Sévaux RG, Alam A, Bleeker-Rovers CP, Bobot M, Cornec-Le Gall E, Gevers TJ, Hassoun Z, Meijer E, Mrug M, Nevens F, Onuchic LF, Pei Y, Piccoli GB, Pirson Y, Rangan GK, Torra R, Visser FW, Jouret F, Kanaan N, Oyen WJ, Suwabe T, Torres VE, and Drenth JP
- Subjects
- Adult, Algorithms, Delphi Technique, Diagnosis, Computer-Assisted, Expert Testimony, Female, Gastroenterology, Humans, Male, Middle Aged, Nephrology, Cysts complications, Hepatitis diagnosis, Hepatitis etiology, Liver Diseases complications, Nephritis diagnosis, Nephritis etiology, Polycystic Kidney, Autosomal Dominant complications
- Abstract
Background: Cyst infection is one of the complications of autosomal dominant polycystic kidney disease and polycystic liver disease. The diagnosis is typically made on a mix of clinical, laboratory and imaging abnormalities but the importance of individual items is uncertain. We aimed to perform a Delphi survey amongst physicians to achieve consensus on diagnostic criteria., Methods: We retrieved diagnostic items from the literature and conducted physician and patient interviews. All items were combined to create the online questionnaire. Participants rated each item during 3 consecutive rounds. Items were rated for diagnostic helpfulness for hepatic and renal cyst infection on a 9-point scale with anchors, from extremely unimportant (n = 1) to extremely important (n = 9). We determined consensus with the disagreement index. The median rating of each item was calculated and categorized into inappropriate (≤3.4), uncertain (3.5-6.4) or appropriate (≥6.5). By combining all items that reached an appropriate consensus rating, we developed a diagnostic algorithm based on expert consensus., Results: We invited 58 physicians to participate in the survey. In total, 35 (60%) responded to round 1 of which 91% (n = 32) and 86% (n = 30) responded to round 2 and 3, respectively. The final panel included 23 nephrologists, 5 hepatologists, a nuclear medicine specialist and an infectious disease physician from 11 countries (male 67%, mean age 47 ± 11 years, median clinical experience 21 years). The panel rated the diagnostic helpfulness of 59 potential items. Ultimately, 22 hepatic and 26 renal items were rated appropriate, including positive blood cultures and fluorodeoxyglucose positron-emission CT imaging. Ultrasonography and absence of intracystic bleeding were amongst those deemed uncertain or inappropriate. Subsequently, by combining items rated appropriate, we developed a clinical tool to diagnose hepatic and renal cyst infection., Conclusions: We identified diagnostic items for hepatic and renal cyst infection and developed an expert-based diagnostic algorithm, which may aid physicians in the diagnostic work-up. A prospective study is necessary to validate this algorithm., (© 2016 S. Karger AG, Basel.)
- Published
- 2016
- Full Text
- View/download PDF
32. Clinical Correlates of Mass Effect in Autosomal Dominant Polycystic Kidney Disease.
- Author
-
Kim H, Park HC, Ryu H, Kim K, Kim HS, Oh KH, Yu SJ, Chung JW, Cho JY, Kim SH, Cheong HI, Lee K, Park JH, Pei Y, Hwang YH, and Ahn C
- Subjects
- Cysts epidemiology, Female, Follow-Up Studies, Humans, Liver Diseases epidemiology, Male, Middle Aged, Organ Size, Polycystic Kidney, Autosomal Dominant epidemiology, Prevalence, Prognosis, Republic of Korea epidemiology, Retrospective Studies, Cysts complications, Cysts pathology, Liver Diseases complications, Liver Diseases pathology, Polycystic Kidney, Autosomal Dominant etiology, Polycystic Kidney, Autosomal Dominant pathology
- Abstract
Mass effect from polycystic kidney and liver enlargement can result in significant clinical complications and symptoms in autosomal dominant polycystic kidney disease (ADPKD). In this single-center study, we examined the correlation of height-adjusted total liver volume (htTLV) and total kidney volume (htTKV) by CT imaging with hepatic complications (n = 461) and abdominal symptoms (n = 253) in patients with ADPKD. "Mass-effect" complications were assessed by review of medical records and abdominal symptoms, by a standardized research questionnaire. Overall, 91.8% of patients had 4 or more liver cysts on CT scans. Polycystic liver disease (PLD) was classified as none or mild (htTLV < 1,600 mL/m); moderate (1,600 ≤ htTLV <3,200 mL/m); and severe (htTLV ≥ 3,200 mL/m). The prevalence of moderate and severe PLD in our patient cohort was 11.7% (n = 54/461) and 4.8% (n = 22/461), respectively, with a female predominance in both the moderate (61.1%) and severe (95.5%) PLD groups. Pressure-related complications such as leg edema (20.4%), ascites (16.6%), and hernia (3.6%) were common, and patients with moderate to severe PLD exhibited a 6-fold increased risk (compared to no or mild PLD) for these complications in multivariate analysis. Similarly, abdominal symptoms including back pain (58.8%), flank pain (53.1%), abdominal fullness (46.5%), and dyspnea/chest-discomfort (44.3%) were very common, and patients with moderate to severe PLD exhibited a 5-fold increased risk for these symptoms. Moderate to severe PLD is a common and clinically important problem in ~16% of patients with ADPKD who may benefit from referral to specialized centers for further management.
- Published
- 2015
- Full Text
- View/download PDF
33. Autosomal-dominant polycystic kidney disease (ADPKD): executive summary from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference.
- Author
-
Chapman AB, Devuyst O, Eckardt KU, Gansevoort RT, Harris T, Horie S, Kasiske BL, Odland D, Pei Y, Perrone RD, Pirson Y, Schrier RW, Torra R, Torres VE, Watnick T, and Wheeler DC
- Subjects
- Consensus, Disease Progression, Humans, Hypertension complications, Hypertension diagnosis, Hypertension therapy, Kidney Failure, Chronic etiology, Kidney Failure, Chronic prevention & control, Polycystic Kidney, Autosomal Dominant complications, Polycystic Kidney, Autosomal Dominant physiopathology, Kidney Failure, Chronic therapy, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant therapy
- Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) affects up to 12 million individuals and is the fourth most common cause for renal replacement therapy worldwide. There have been many recent advances in the understanding of its molecular genetics and biology, and in the diagnosis and management of its manifestations. Yet, diagnosis, evaluation, prevention, and treatment vary widely and there are no broadly accepted practice guidelines. Barriers to translation of basic science breakthroughs to clinical care exist, with considerable heterogeneity across countries. The Kidney Disease: Improving Global Outcomes Controversies Conference on ADPKD brought together a panel of multidisciplinary clinical expertise and engaged patients to identify areas of consensus, gaps in knowledge, and research and health-care priorities related to diagnosis; monitoring of kidney disease progression; management of hypertension, renal function decline and complications; end-stage renal disease; extrarenal complications; and practical integrated patient support. These are summarized in this review.
- Published
- 2015
- Full Text
- View/download PDF
34. Imaging-based diagnosis of autosomal dominant polycystic kidney disease.
- Author
-
Pei Y, Hwang YH, Conklin J, Sundsbak JL, Heyer CM, Chan W, Wang K, He N, Rattansingh A, Atri M, Harris PC, and Haider MA
- Subjects
- Adolescent, Adult, Case-Control Studies, Female, Humans, Magnetic Resonance Imaging, Male, Ultrasonography, Young Adult, Polycystic Kidney, Autosomal Dominant diagnostic imaging
- Abstract
The clinical use of conventional ultrasonography (US) in autosomal dominant polycystic kidney disease (ADPKD) is currently limited by reduced diagnostic sensitivity, especially in at-risk subjects younger than 30 years of age. In this single-center prospective study, we compared the diagnostic performance of MRI with that of high-resolution (HR) US in 126 subjects ages 16-40 years born with a 50% risk of ADPKD who underwent both these renal imaging studies and comprehensive PKD1 and PKD2 mutation screening. Concurrently, 45 healthy control subjects without a family history of ADPKD completed the same imaging protocol. We analyzed 110 at-risk subjects whose disease status was unequivocally defined by molecular testing and 45 unaffected healthy control subjects. Using a total of >10 cysts as a test criterion in subjects younger than 30 years of age, we found that MRI provided both a sensitivity and specificity of 100%. Comparison of our results from HR US with those from a previous study of conventional US using the test criterion of a total of three or more cysts found a higher diagnostic sensitivity (approximately 97% versus approximately 82%) with a slightly decreased specificity (approximately 98% versus 100%) in this study. Similar results were obtained in test subjects between the ages of 30 and 40 years old. These results suggest that MRI is highly sensitive and specific for diagnosis of ADPKD. HR US has the potential to rival the diagnostic performance of MRI but is both center- and operator-dependent., (Copyright © 2015 by the American Society of Nephrology.)
- Published
- 2015
- Full Text
- View/download PDF
35. The cleaved cytoplasmic tail of polycystin-1 regulates Src-dependent STAT3 activation.
- Author
-
Talbot JJ, Song X, Wang X, Rinschen MM, Doerr N, LaRiviere WB, Schermer B, Pei YP, Torres VE, and Weimbs T
- Subjects
- Animals, Cell Culture Techniques, Cyclic AMP genetics, Cyclic AMP metabolism, Disease Models, Animal, Dogs, ErbB Receptors physiology, Mice, Polycystic Kidney, Autosomal Dominant metabolism, Polycystic Kidney, Autosomal Dominant pathology, Protein-Tyrosine Kinases physiology, RNA, Messenger metabolism, Rats, Signal Transduction physiology, Suppressor of Cytokine Signaling Proteins genetics, Suppressor of Cytokine Signaling Proteins metabolism, Polycystic Kidney, Autosomal Dominant etiology, STAT3 Transcription Factor physiology, TRPP Cation Channels physiology
- Abstract
Polycystin-1 (PC1) mutations result in proliferative renal cyst growth and progression to renal failure in autosomal dominant polycystic kidney disease (ADPKD). The transcription factor STAT3 (signal transducer and activator of transcription 3) was shown to be activated in cyst-lining cells in ADPKD and PKD mouse models and may drive renal cyst growth, but the mechanisms leading to persistent STAT3 activation are unknown. A proteolytic fragment of PC1 corresponding to the cytoplasmic tail, PC1-p30, is overexpressed in ADPKD. Here, we show that PC1-p30 interacts with the nonreceptor tyrosine kinase Src, resulting in Src-dependent activation of STAT3 by tyrosine phosphorylation. The PC1-p30-mediated activation of Src/STAT3 was independent of JAK family kinases and insensitive to the STAT3 inhibitor suppressor of cytokine signaling 3. Signaling by the EGF receptor (EGFR) or cAMP amplified the activation of Src/STAT3 by PC1-p30. Expression of PC1-p30 changed the cellular response to cAMP signaling. In the absence of PC1-p30, cAMP dampened EGFR- or IL-6-dependent activation of STAT3; in the presence of PC1-p30, cAMP amplified Src-dependent activation of STAT3. In the polycystic kidney (PCK) rat model, activation of STAT3 in renal cystic cells depended on vasopressin receptor 2 (V2R) signaling, which increased cAMP levels. Genetic inhibition of vasopressin expression or treatment with a pharmacologic V2R inhibitor strongly suppressed STAT3 activation and reduced renal cyst growth. These results suggest that PC1, via its cleaved cytoplasmic tail, integrates signaling inputs from EGFR and cAMP, resulting in Src-dependent activation of STAT3 and a proliferative response., (Copyright © 2014 by the American Society of Nephrology.)
- Published
- 2014
- Full Text
- View/download PDF
36. A missense mutation in PKD1 attenuates the severity of renal disease.
- Author
-
Pei Y, Lan Z, Wang K, Garcia-Gonzalez M, He N, Dicks E, Parfrey P, Germino G, and Watnick T
- Subjects
- Animals, Apoptosis genetics, Cell Proliferation, Cells, Cultured, Cysts genetics, Dogs, Genotype, Humans, Mutation, Missense, Pedigree, Phenotype, Severity of Illness Index, Polycystic Kidney, Autosomal Dominant genetics, TRPP Cation Channels genetics
- Abstract
Mutations of PKD1 and PKD2 account for most cases of autosomal dominant polycystic kidney disease (ADPKD). Compared with PKD2, patients with PKD1 typically have more severe renal disease. Here, we report a follow-up study of a unique multigeneration family with bilineal ADPKD (NFL10) in which a PKD1 disease haplotype and a PKD2 (L736X) mutation co-segregated with 18 and 14 affected individuals, respectively. In our updated genotype-phenotype analysis of the family, we found that PKD1-affected individuals had uniformly mild renal disease similar to the PKD2-affected individuals. By sequencing all the exons and splice junctions of PKD1, we identified two missense mutations (Y528C and R1942H) from a PKD1-affected individual. Although both variants were predicted to be damaging to the mutant protein, only Y528C co-segregated with all of the PKD1-affected individuals in NFL10. Studies in MDCK cells stably expressing wild-type and mutant forms of PKD found that cell lines expressing the Y528C variant formed cysts in culture and displayed increased rates of growth and apoptosis. Thus, Y528C functions as a hypomorphic PKD1 allele. These findings have important implications for pathogenic mechanisms and molecular diagnostics of ADPKD.
- Published
- 2012
- Full Text
- View/download PDF
37. Practical genetics for autosomal dominant polycystic kidney disease.
- Author
-
Pei Y
- Subjects
- Animals, Humans, Forecasting, Genetic Predisposition to Disease genetics, Genetic Testing methods, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common mendelian disorder of the kidney and accounts for ~5% of end-stage renal disease in North America. It is characterized by focal development of renal cysts which increase in number and size with age. Mutations of PKD1 and PKD2 account for most cases. Although the clinical manifestations of both gene types overlap completely, PKD1 is associated with more severe disease than PKD2, with larger kidneys and earlier onset of end-stage renal disease. Furthermore, marked within-family renal disease variability is well documented in ADPKD and suggests a strong modifier effect from as yet unknown genetic and environmental factors. In turn, the significant inter- and intra-familial renal disease variability poses a challenge for diagnosis and genetic counseling. In general, renal ultrasonography is commonly used for the diagnosis, and age-dependent criteria have been defined for subjects at risk of PKD1. However, the utility of the PKD1 ultrasound criteria in the clinical setting is unclear since their performance characteristics have not been defined for the milder PKD2 and the gene type for most test subjects is unknown. Recently, highly predictive ultrasound diagnostic criteria have been derived for at-risk subjects of unknown gene type. Additionally, both DNA linkage and gene-based direct sequencing are available for the diagnosis of ADPKD, especially in subjects with equivocal imaging results, a negative or indeterminate family history, or in younger at-risk individuals being evaluated as potential living related kidney donor. This review will highlight the utility and limitations of clinical predictors of gene types, imaging- and molecular-based diagnostic tests, and present an integrated approach for evaluating individuals suspected to have ADPKD., (Copyright © 2010 S. Karger AG, Basel.)
- Published
- 2011
- Full Text
- View/download PDF
38. Genetic variation of DKK3 may modify renal disease severity in ADPKD.
- Author
-
Liu M, Shi S, Senthilnathan S, Yu J, Wu E, Bergmann C, Zerres K, Bogdanova N, Coto E, Deltas C, Pierides A, Demetriou K, Devuyst O, Gitomer B, Laakso M, Lumiaho A, Lamnissou K, Magistroni R, Parfrey P, Breuning M, Peters DJ, Torra R, Winearls CG, Torres VE, Harris PC, Paterson AD, and Pei Y
- Subjects
- Adaptor Proteins, Signal Transducing, Adult, Chemokines, Cohort Studies, Genotype, Humans, Middle Aged, Mutation, Phenotype, TRPP Cation Channels genetics, Intercellular Signaling Peptides and Proteins genetics, Polycystic Kidney, Autosomal Dominant genetics, Polymorphism, Single Nucleotide
- Abstract
Significant variation in the course of autosomal dominant polycystic kidney disease ( ADPKD) within families suggests the presence of effect modifiers. Recent studies of the variation within families harboring PKD1 mutations indicate that genetic background may account for 32 to 42% of the variance in estimated GFR (eGFR) before ESRD and 43 to 78% of the variance in age at ESRD onset, but the genetic modifiers are unknown. Here, we conducted a high-throughput single-nucleotide polymorphism (SNP) genotyping association study of 173 biological candidate genes in 794 white patients from 227 families with PKD1. We analyzed two primary outcomes: (1) eGFR and (2) time to ESRD (renal survival). For both outcomes, we used multidimensional scaling to correct for population structure and generalized estimating equations to account for the relatedness among individuals within the same family. We found suggestive associations between each of 12 SNPs and at least one of the renal outcomes. We genotyped these SNPs in a second set of 472 white patients from 229 families with PKD1 and performed a joint analysis on both cohorts. Three SNPs continued to show suggestive/significant association with eGFR at the Dickkopf 3 (DKK3) gene locus; no SNPs significantly associated with renal survival. DKK3 antagonizes Wnt/beta-catenin signaling, which may modulate renal cyst growth. Pending replication, our study suggests that genetic variation of DKK3 may modify severity of ADPKD resulting from PKD1 mutations.
- Published
- 2010
- Full Text
- View/download PDF
39. Diagnosis of autosomal-dominant polycystic kidney disease: an integrated approach.
- Author
-
Barua M and Pei Y
- Subjects
- Adolescent, Adult, Age Factors, Algorithms, Genotype, Humans, Middle Aged, Pedigree, Ultrasonography, Young Adult, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Polycystic Kidney, Autosomal Dominant genetics, TRPP Cation Channels genetics
- Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is the most common Mendelian disorder of the kidney and accounts for approximately 5% of end-stage renal disease in developed countries. It is characterized by focal and sporadic development of renal cysts that increase in number and size with age. Mutations of 2 genes (ie, PKD1 and PKD2) account for most of the cases. Although the clinical manifestations of both gene types overlap completely, PKD1 is associated with more severe disease than PKD2, with bigger kidneys and earlier onset of end-stage renal disease. In general, the diagnosis of ADPKD is commonly made by renal ultrasonography. Age-dependent ultrasound criteria have been established for both diagnosis and disease exclusion in subjects at risk of PKD1. However, the utility of these criteria in the clinic setting is unclear because their performance characteristics have not been defined for the milder PKD2 and the gene type for most test subjects is unknown. Recently, highly predictive ultrasound diagnostic criteria have been derived for at-risk subjects of unknown gene type. In addition, molecular genetic testing is now available for the diagnosis of ADPKD, especially in subjects with equivocal imaging results, with a negative or indeterminate family history, or in younger at-risk individuals with a negative ultrasound study being evaluated as potential living-related kidney donor. Here, we review the clinical utilities and limitations of these imaging- and molecular-based diagnostic tests, and outline our approach for the evaluation of individuals suspected to have ADPKD., (Copyright 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
40. Diagnosis and screening of autosomal dominant polycystic kidney disease.
- Author
-
Pei Y and Watnick T
- Subjects
- Diagnosis, Differential, Humans, Polycystic Kidney, Autosomal Dominant epidemiology, Risk Factors, Genetic Testing, Mass Screening, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of kidney failure and accounts for approximately 5% of ESRD population in the United States. The disorder is characterized by the focal and sporadic development of renal cysts, which increase in size and number with age. Mutations of PKD1 and PKD2 account for most of the cases. Although the clinical manifestations of both gene types overlap completely, PKD1 is associated with more severe disease than PKD2, with larger kidneys and earlier onset of ESRD. In general, renal ultrasonography is commonly used for the diagnosis of ADPKD, and age-dependent criteria have been defined for subjects at risk of PKD1. However, the utility of the PKD1 ultrasound criteria in the clinic setting is unclear because their performance characteristics have not been defined for the milder PKD2 and the gene type for most test subjects is unknown. Recently, highly predictive ultrasound diagnostic criteria have been derived for at-risk subjects of unknown gene type. Additionally, both DNA linkage or gene-based direct sequencing are now available for the diagnosis of ADPKD, especially in subjects with equivocal imaging results, subjects with a negative or indeterminate family history, or in younger at-risk individuals being evaluated as potential living-related kidney donors. Here, we review the clinical utilities and limitations of both imaging- and molecular-based diagnostic tests and outline our approach for the evaluation of individuals suspected to have ADPKD., (2010 National Kidney Foundation, Inc. Published by Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
41. Autosomal dominant polycystic kidney disease.
- Author
-
Pei Y and Watnick T
- Subjects
- Animals, Disease Models, Animal, Humans, Genotype, Polycystic Kidney, Autosomal Dominant epidemiology, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant physiopathology
- Published
- 2010
- Full Text
- View/download PDF
42. Family history of renal disease severity predicts the mutated gene in ADPKD.
- Author
-
Barua M, Cil O, Paterson AD, Wang K, He N, Dicks E, Parfrey P, and Pei Y
- Subjects
- Adolescent, Adult, Age of Onset, Aged, Aged, 80 and over, Child, Child, Preschool, DNA Mutational Analysis, Humans, Kidney Failure, Chronic epidemiology, Middle Aged, Ontario epidemiology, Polycystic Kidney, Autosomal Dominant complications, Young Adult, Kidney Failure, Chronic etiology, Polycystic Kidney, Autosomal Dominant genetics, TRPP Cation Channels genetics
- Abstract
Mutations of PKD1 and PKD2 account for 85 and 15% of cases of autosomal dominant polycystic kidney disease (ADPKD), respectively. Clinically, PKD1 is more severe than PKD2, with a median age at ESRD of 53.4 versus 72.7 yr. In this study, we explored whether a family history of renal disease severity predicts the mutated gene in ADPKD. We examined the renal function (estimated GFR and age at ESRD) of 484 affected members from 90 families who had ADPKD and whose underlying genotype was known. We found that the presence of at least one affected family member who developed ESRD at age < or =55 was highly predictive of a PKD1 mutation (positive predictive value 100%; sensitivity 72%). In contrast, the presence of at least one affected family member who continued to have sufficient renal function or developed ESRD at age >70 was highly predictive of a PKD2 mutation (positive predictive value 100%; sensitivity 74%). These data suggest that close attention to the family history of renal disease severity in ADPKD may provide a simple means of predicting the mutated gene, which has prognostic implications.
- Published
- 2009
- Full Text
- View/download PDF
43. Systems biology of autosomal dominant polycystic kidney disease (ADPKD): computational identification of gene expression pathways and integrated regulatory networks.
- Author
-
Song X, Di Giovanni V, He N, Wang K, Ingram A, Rosenblum ND, and Pei Y
- Subjects
- Cysts genetics, Cysts metabolism, Humans, Oligonucleotide Array Sequence Analysis, Polycystic Kidney, Autosomal Dominant metabolism, Signal Transduction, Systems Biology, Transcription Factors genetics, Transcription Factors metabolism, Gene Expression Profiling, Gene Regulatory Networks, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
To elucidate the molecular pathways that modulate renal cyst growth in ADPKD, we performed global gene profiling on cysts of different size (<1 ml, n = 5; 10-20 ml, n = 5; >50 ml, n = 3) and minimally cystic tissue (MCT, n = 5) from five PKD1 human polycystic kidneys using Affymetrix HG-U133 Plus 2.0 arrays. We used gene set enrichment analysis to identify overrepresented signaling pathways and key transcription factors (TFs) between cysts and MCT. We found down-regulation of kidney epithelial restricted genes (e.g. nephron segment-specific markers and cilia-associated cystic genes such as HNF1B, PKHD1, IFT88 and CYS1) in the renal cysts. On the other hand, PKD1 cysts displayed a rich profile of gene sets associated with renal development, mitogen-mediated proliferation, cell cycle progression, epithelial-mesenchymal transition, hypoxia, aging and immune/inflammatory responses. Notably, our data suggest that up-regulation of Wnt/beta-catenin, pleiotropic growth factor/receptor tyrosine kinase (e.g. IGF/IGF1R, FGF/FGFR, EGF/EGFR, VEGF/VEGFR), G-protein-coupled receptor (e.g. PTGER2) signaling was associated with renal cystic growth. By integrating these pathways with a number of dysregulated networks of TFs (e.g. SRF, MYC, E2F1, CREB1, LEF1, TCF7, HNF1B/ HNF1A and HNF4A), our data suggest that epithelial dedifferentiation accompanied by aberrant activation and cross-talk of specific signaling pathways may be required for PKD1 cyst growth and disease progression. Pharmacological modulation of some of these signaling pathways may provide a potential therapeutic strategy for ADPKD.
- Published
- 2009
- Full Text
- View/download PDF
44. Evidence for pathogenicity of atypical splice mutations in autosomal dominant polycystic kidney disease.
- Author
-
Wang K, Zhao X, Chan S, Cil O, He N, Song X, Paterson AD, and Pei Y
- Subjects
- Adult, Codon, Nonsense, Computational Biology, DNA Mutational Analysis, Exons, Female, Genetic Predisposition to Disease, Genetic Testing, Haplotypes, Humans, Male, Microsatellite Repeats, Middle Aged, Phenotype, Polycystic Kidney, Autosomal Dominant diagnostic imaging, Reverse Transcriptase Polymerase Chain Reaction, Ultrasonography, Kidney diagnostic imaging, Mutation, Polycystic Kidney, Autosomal Dominant genetics, RNA Splice Sites, TRPP Cation Channels genetics
- Abstract
Background and Objectives: Mutation-based molecular diagnostics of autosomal dominant polycystic kidney disease (ADPKD) is complicated by locus and allelic heterogeneity, large multi-exon gene structure and duplication in PKD1, and a high level of unclassified variants. Comprehensive screening of PKD1 and PKD2 by two recent studies have shown that atypical splice mutations account for 3.5% to 5% of ADPKD. We evaluated the role of bioinformatic prediction of atypical splice mutations and determined the pathogenicity of an atypical PKD2 splice variant from a multiplex ADPKD (TOR101) family., Design, Setting, Participants, & Measurements: Using PubMed, we identified 17 atypical PKD1 and PKD2 splice mutations. We found that bioinformatics analysis was often useful for evaluating the pathogenicity of these mutations, although RT-PCR is needed to provide the definitive proof., Results: Sequencing of both PKD1 and PKD2 in an affected subject of TOR101 failed to identify a definite mutation, but revealed several UCVs, including an atypical PKD2 splice variant. Linkage analysis with microsatellite markers indicated that TOR101 was PKD2-linked and IVS8 + 5G-->A was shown to cosegregate only with affected subjects. RT-PCR of leukocyte mRNA from an affected subject using primers from exons 7 and 9 revealed six splice variants that resulted from activation of different combinations of donor and acceptor cryptic splice sites, all terminating with premature stop codons., Conclusions: The data provide strong evidence that IVS8 + 5G-->A is a pathogenic mutation for PKD2. This case highlights the importance of functional analysis of UCVs.
- Published
- 2009
- Full Text
- View/download PDF
45. Unified criteria for ultrasonographic diagnosis of ADPKD.
- Author
-
Pei Y, Obaji J, Dupuis A, Paterson AD, Magistroni R, Dicks E, Parfrey P, Cramer B, Coto E, Torra R, San Millan JL, Gibson R, Breuning M, Peters D, and Ravine D
- Subjects
- Adolescent, Adult, Genotype, Humans, Middle Aged, Mutation, Polycystic Kidney, Autosomal Dominant genetics, Ultrasonography, Polycystic Kidney, Autosomal Dominant diagnostic imaging, TRPP Cation Channels genetics
- Abstract
Individuals who are at risk for autosomal dominant polycystic kidney disease are often screened by ultrasound using diagnostic criteria derived from individuals with mutations in PKD1. Families with mutations in PKD2 typically have less severe disease, suggesting a potential need for different diagnostic criteria. In this study, 577 and 371 at-risk individuals from 58 PKD1 and 39 PKD2 families, respectively, were assessed by renal ultrasound and molecular genotyping. Using sensitivity data derived from genetically affected individuals and specificity data derived from genetically unaffected individuals, various diagnostic criteria were compared. In addition, data sets were created to simulate the PKD1 and PKD2 case mix expected in practice to evaluate the performance of diagnostic criteria for families of unknown genotype. The diagnostic criteria currently in use performed suboptimally for individuals with mutations in PKD2 as a result of reduced test sensitivity. In families of unknown genotype, the presence of three or more (unilateral or bilateral) renal cysts is sufficient for establishing the diagnosis in individuals aged 15 to 39 y, two or more cysts in each kidney is sufficient for individuals aged 40 to 59 y, and four or more cysts in each kidney is required for individuals > or = 60 yr. Conversely, fewer than two renal cysts in at-risk individuals aged > or = 40 yr is sufficient to exclude the disease. These unified diagnostic criteria will be useful for testing individuals who are at risk for autosomal dominant polycystic kidney disease in the usual clinical setting in which molecular genotyping is seldom performed.
- Published
- 2009
- Full Text
- View/download PDF
46. Presence of de novo mutations in autosomal dominant polycystic kidney disease patients without family history.
- Author
-
Reed B, McFann K, Kimberling WJ, Pei Y, Gabow PA, Christopher K, Petersen E, Kelleher C, Fain PR, Johnson A, and Schrier RW
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Genetic Testing, Humans, Male, Middle Aged, TRPP Cation Channels genetics, Young Adult, Mutation, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Background: At the University of Colorado Health Sciences Center, on detailed questioning, approximately 10% of patients with autosomal dominant polycystic kidney disease (ADPKD) gave no family history of ADPKD. There are several explanations for this observation, including occurrence of a de novo pathogenic sequence variant or extreme phenotypic variability. To confirm de novo sequence variants, we have undertaken clinical and genetic screening of affected offspring and their parents., Study Design: Case series., Setting & Participants: 24 patients with a well-documented ADPKD phenotype and no family history of polycystic kidney disease (PKD) and both parents of each patient., Outcome: Presence or absence of PKD1 or PKD2 pathogenic sequence variants in parents of affected offspring., Measurements: Abdominal ultrasound of affected offspring and their parents for ADPKD diagnosis. Parentage testing by genotyping. Complete screening of PKD1 and PKD2 genes by using genomic DNA from affected offspring; analysis of genomic DNA from both parents to confirm the absence or presence of all DNA variants found., Results: A positive diagnosis of ADPKD by means of ultrasound or genetic screening was made in 1 parent of 4 patients (17%). No PKD1 or PKD2 pathogenic sequence variants were identified in 10 patients (42%), whereas possible pathological DNA variants were identified in 4 patients (17%) and 1 of their respective parents. Parentage was confirmed in the remaining 6 patients (25%), and de novo sequence variants were documented., Limitations: Size of patient group. No direct examination of RNA., Conclusion: Causes other than de novo pathogenic sequence variants may explain the negative family history of ADPKD in certain families.
- Published
- 2008
- Full Text
- View/download PDF
47. Polycystin-1 C-terminal tail associates with beta-catenin and inhibits canonical Wnt signaling.
- Author
-
Lal M, Song X, Pluznick JL, Di Giovanni V, Merrick DM, Rosenblum ND, Chauvet V, Gottardi CJ, Pei Y, and Caplan MJ
- Subjects
- Animals, Binding Sites, CHO Cells, Cell Line, Cell Nucleus metabolism, Cricetinae, Cricetulus, Gene Expression Profiling, Gene Expression Regulation, Humans, Ligands, Oligonucleotide Array Sequence Analysis, Peptide Fragments chemistry, Peptide Fragments genetics, Peptide Fragments metabolism, Polycystic Kidney, Autosomal Dominant etiology, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Signal Transduction, Systems Biology, TCF Transcription Factors genetics, Transfection, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant metabolism, TRPP Cation Channels chemistry, TRPP Cation Channels metabolism, Wnt Proteins metabolism, beta Catenin metabolism
- Abstract
Polycystin-1 (PC1), the product of the PKD1 gene mutated in the majority of autosomal dominant polycystic kidney disease (ADPKD) cases, undergoes a cleavage resulting in the intracellular release of its C-terminal tail (CTT). Here, we demonstrate that the PC1 CTT co-localizes with and binds to beta-catenin in the nucleus. This interaction requires a nuclear localization motif present in the PC1 CTT as well as the N-terminal portion of beta-catenin. The PC1 CTT inhibits the ability of both beta-catenin and Wnt ligands to activate T-cell factor (TCF)-dependent gene transcription, a major effector of the canonical Wnt signaling pathway. The PC1 CTT may produce this effect by reducing the apparent affinity of the interaction between beta-catenin and the TCF protein. DNA microarray analysis reveals that the canonical Wnt signaling pathway is activated in ADPKD patient cysts. Our results suggest a novel mechanism through which PC1 cleavage may impact upon Wnt-dependent signaling and thereby modulate both developmental processes and cystogenesis.
- Published
- 2008
- Full Text
- View/download PDF
48. Molecular diagnostics in autosomal dominant polycystic kidney disease: utility and limitations.
- Author
-
Zhao X, Paterson AD, Zahirieh A, He N, Wang K, and Pei Y
- Subjects
- Adolescent, Adult, Aged, DNA Mutational Analysis methods, DNA Mutational Analysis standards, Family Health, Female, Genetic Linkage, Haplotypes, Humans, Male, Middle Aged, Pedigree, TRPP Cation Channels, Genetic Testing methods, Genetic Testing standards, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Background and Objectives: Gene-based mutation screening is now available and has the potential to provide diagnostic confirmation or exclusion of autosomal dominant polycystic kidney disease. This study illustrates its utility and limitations in the clinical setting., Design, Setting, Participants, & Measurements: Using a molecular diagnostic service, genomic DNA of one affected individual from each study family was screened for pathologic PKD1 and PKD2 mutations. Bidirectional sequencing was performed to identify sequence variants in all exons and splice junctions of both genes and to confirm the specific mutations in other family members. In two multiplex families, microsatellite markers were genotyped at both PDK1 and PKD2 loci, and pair-wise and multipoint linkage analysis was performed., Results: Three of five probands studied were referred for assessment of renal cystic disease without a family history of autosomal dominant polycystic kidney disease, and two others were younger at-risk members of families with autosomal dominant polycystic kidney disease being evaluated as living-related kidney donors. Gene-based mutation screening identified pathogenic mutations that provided confirmation or exclusion of disease in three probands, but in the other two, only unclassified variants were identified. In one proband in which mutation screening was indeterminate, DNA linkage studies provided strong evidence for disease exclusion., Conclusions: Gene-based mutation screening or DNA linkage analysis should be considered in individuals in whom the diagnosis of autosomal dominant polycystic kidney disease is uncertain because of a lack of family history or equivocal imaging results and in younger at-risk individuals who are being evaluated as living-related kidney donors.
- Published
- 2008
- Full Text
- View/download PDF
49. Evaluating the clinical utility of a molecular genetic test for polycystic kidney disease.
- Author
-
Garcia-Gonzalez MA, Jones JG, Allen SK, Palatucci CM, Batish SD, Seltzer WK, Lan Z, Allen E, Qian F, Lens XM, Pei Y, Germino GG, and Watnick TJ
- Subjects
- Adult, Age of Onset, Aged, Amino Acid Sequence, Codon, Nonsense, DNA Mutational Analysis, Female, Frameshift Mutation genetics, Gene Deletion, Genetic Variation, Genotype, Humans, Male, Middle Aged, Molecular Diagnostic Techniques, Molecular Sequence Data, Polymorphism, Genetic, RNA Splice Sites, Sequence Homology, Amino Acid, TRPP Cation Channels analysis, Genetic Testing statistics & numerical data, Polycystic Kidney, Autosomal Dominant diagnosis, Polycystic Kidney, Autosomal Dominant genetics
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is estimated to affect 1/600-1/1000 individuals worldwide. The disease is characterized by age dependent renal cyst formation that results in kidney failure during adulthood. Although ultrasound imaging may be an adequate diagnostic tool in at risk individuals older than 30, this modality may not be sufficiently sensitive in younger individuals or for those from PKD2 families who have milder disease. DNA based assays may be indicated in certain clinical situations where imaging cannot provide a definitive clinical diagnosis. The goal of this study was to evaluate the utility of direct DNA analysis in a test sample of 82 individuals who were judged to have polycystic kidney disease by standard clinical criteria. The samples were analyzed using a commercially available assay that employs sequencing of both genes responsible for the disorder. Definite disease causing mutations were identified in 34 (approximately 42%) study participants. An additional 30 (approximately 37%) subjects had either in frame insertions/deletions, non-canonical splice site alterations or a combination of missense changes that were also judged likely to be pathogenic. We noted striking sequence variability in the PKD1 gene, with a mean of 13.1 variants per participant (range 0-60). Our results and analysis highlight the complexity of assessing the pathogenicity of missense variants particularly when individuals have multiple amino acid substitutions. We conclude that a significant fraction of ADPKD mutations are caused by amino acid substitutions that need to be interpreted carefully when utilized in clinical decision-making.
- Published
- 2007
- Full Text
- View/download PDF
50. Diagnostic approach in autosomal dominant polycystic kidney disease.
- Author
-
Pei Y
- Subjects
- Age Factors, Algorithms, Decision Trees, Diagnosis, Differential, Genetic Counseling, Genetic Predisposition to Disease, Humans, Kidney Failure, Chronic genetics, Kidney Failure, Chronic pathology, Magnetic Resonance Imaging, Mutation, Pedigree, Polycystic Kidney, Autosomal Dominant complications, Polycystic Kidney, Autosomal Dominant genetics, Polycystic Kidney, Autosomal Dominant pathology, Predictive Value of Tests, Reproducibility of Results, Risk Assessment, Sensitivity and Specificity, Severity of Illness Index, TRPP Cation Channels, Tomography, X-Ray Computed, Ultrasonography methods, Diagnostic Imaging methods, Genetic Testing, Kidney Failure, Chronic etiology, Molecular Diagnostic Techniques, Polycystic Kidney, Autosomal Dominant diagnosis
- Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common Mendelian disorder of the kidney and affects all racial groups worldwide. It is characterized by focal development of renal and extrarenal cysts in an age-dependent manner. Typically, only a few renal cysts are detected in most affected individuals before 30 yr of age. However, by the fifth decade of life, hundreds to thousands of renal cysts will be found in the majority of patients. ADPKD is genetically heterogeneous. Mutations of two genes, PKD1 and PKD2, account for approximately 85 and 15% of cases, respectively. Although the clinical manifestations of these two genotypes overlap completely, patients with PKD1 have much more severe renal disease compared with those with PKD2, as evidenced by their ESRD occurring approximately 15 yr earlier. Renal ultrasonography commonly is used for the assessment of ADPKD, and age-dependent ultrasound diagnostic criteria with high sensitivity and specificity have been established for individuals who are born with 50% risk for PKD1. Although these diagnostic criteria are used widely for genetic counseling and for the evaluation of at-risk individuals as living-related kidney donors to their affected relatives, their application to individuals who are at risk for PKD2 or have undefined genotype needs to be refined further. Molecular genetic testing is available for ADPKD and may be useful for evaluation of at-risk individuals with equivocal imaging results, younger at-risk individuals as a living-related kidney donor, and individuals with atypical or de novo renal cystic disease.
- Published
- 2006
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.