1
|
Ha K, Mundt-Machado N, Bisignano P, Pinedo A, Raleigh DR, Loeb G, Reiter JF, Cao E, Delling M. Cilia-enriched oxysterol 7β,27-DHC is required for polycystin ion channel activation. Nat Commun 2024; 15:6468. [PMID: 39085216 PMCID: PMC11291729 DOI: 10.1038/s41467-024-50318-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 06/28/2024] [Indexed: 08/02/2024] Open
Abstract
Polycystin-1 (PC-1) and PC-2 form a heteromeric ion channel complex that is abundantly expressed in primary cilia of renal epithelial cells. This complex functions as a non-selective cation channel, and mutations within the polycystin complex cause autosomal dominant polycystic kidney disease (ADPKD). The spatial and temporal regulation of the polycystin complex within the ciliary membrane remains poorly understood. Using both whole-cell and ciliary patch-clamp recordings, we identify a cilia-enriched oxysterol, 7β,27-dihydroxycholesterol (DHC), that serves as a necessary activator of the polycystin complex. We further identify an oxysterol-binding pocket within PC-2 and showed that mutations within this binding pocket disrupt 7β,27-DHC-dependent polycystin activation. Pharmacologic and genetic inhibition of oxysterol synthesis reduces channel activity in primary cilia. In summary, our findings reveal a regulator of the polycystin complex. This oxysterol-binding pocket in PC-2 may provide a specific target for potential ADPKD therapeutics.
Collapse
Affiliation(s)
- Kodaji Ha
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Nadine Mundt-Machado
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - Paola Bisignano
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Aide Pinedo
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA
| | - David R Raleigh
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriel Loeb
- Department of Medicine, University of California, San Francisco, CA, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Markus Delling
- Department of Physiology, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
2
|
Luo J, Zhang Y, Jayaprakash S, Zhuang L, He J. Cross-Species Insights into Autosomal Dominant Polycystic Kidney Disease: Provide an Alternative View on Research Advancement. Int J Mol Sci 2024; 25:5646. [PMID: 38891834 PMCID: PMC11171680 DOI: 10.3390/ijms25115646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/09/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a prevalent hereditary disorder that affects the kidneys, characterized by the development of an excessive number of fluid-filled cysts of varying sizes in both kidneys. Along with the progression of ADPKD, these enlarged cysts displace normal kidney tissue, often accompanied by interstitial fibrosis and inflammation, and significantly impair renal function, leading to end-stage renal disease. Currently, the precise mechanisms underlying ADPKD remain elusive, and a definitive cure has yet to be discovered. This review delineates the epidemiology, pathological features, and clinical diagnostics of ADPKD or ADPKD-like disease across human populations, as well as companion animals and other domesticated species. A light has been shed on pivotal genes and biological pathways essential for preventing and managing ADPKD, which underscores the importance of cross-species research in addressing this complex condition. Treatment options are currently limited to Tolvaptan, dialysis, or surgical excision of large cysts. However, comparative studies of ADPKD across different species hold promise for unveiling novel insights and therapeutic strategies to combat this disease.
Collapse
Affiliation(s)
- Jianing Luo
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| | - Yuan Zhang
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| | - Sakthidasan Jayaprakash
- Department of Biotechnology, Hindustan Institute of Technology and Science, Tamil Nadu 603103, India;
| | - Lenan Zhuang
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| | - Jin He
- College of Animal Sciences, Zhejiang University, Hangzhou 310027, China; (J.L.); (Y.Z.); (L.Z.)
| |
Collapse
|
3
|
Zhang Y, Xu S, Jin Q, Luo J, Gao C, Jayaprakash S, Wang H, Zhuang L, He J. Establishment of transgenic pigs overexpressing human PKD2-D511V mutant. Front Genet 2022; 13:1059682. [DOI: 10.3389/fgene.2022.1059682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
Numerous missense mutations have been reported in autosomal dominant polycystic kidney disease which is one of the most common renal genetic disorders. The underlying mechanism for cystogenesis is still elusive, partly due to the lack of suitable animal models. Currently, we tried to establish a porcine transgenic model overexpressing human PKD2-D511V (hPKD2-D511V), which is a dominant-negative mutation in the vertebrate in vitro models. A total of six cloned pigs were finally obtained using somatic cell nuclear transfer. However, five with functional hPKD2-D511V died shortly after birth, leaving only one with the dysfunctional transgenic event to survive. Compared with the WT pigs, the demised transgenic pigs had elevated levels of hPKD2 expression at the mRNA and protein levels. Additionally, no renal malformation was observed, indicating that hPKD2-D511V did not alter normal kidney development. RNA-seq analysis also revealed that several ADPKD-related pathways were disturbed when overexpressing hPKD2-D511V. Therefore, our study implies that hPKD2-D511V may be lethal due to the dominant-negative effect. Hence, to dissect how PKD2-D511V drives renal cystogenesis, it is better to choose in vitro or invertebrate models.
Collapse
|
4
|
Cantero MDR, Cantiello HF. Polycystin-2 (TRPP2): Ion channel properties and regulation. Gene 2022; 827:146313. [PMID: 35314260 DOI: 10.1016/j.gene.2022.146313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 01/19/2022] [Accepted: 02/08/2022] [Indexed: 12/01/2022]
Abstract
Polycystin-2 (TRPP2, PKD2, PC2) is the product of the PKD2 gene, whose mutations cause Autosomal Dominant Polycystic Kidney Disease (ADPKD). PC2 belongs to the superfamily of TRP (Transient Receptor Potential) proteins that generally function as Ca2+-permeable nonselective cation channels implicated in Ca2+ signaling. PC2 localizes to various cell domains with distinct functions that likely depend on interactions with specific channel partners. Functions include receptor-operated, nonselective cation channel activity in the plasma membrane, intracellular Ca2+ release channel activity in the endoplasmic reticulum (ER), and mechanosensitive channel activity in the primary cilium of renal epithelial cells. Here we summarize our current understanding of the properties of PC2 and how other transmembrane and cytosolic proteins modulate this activity, providing functional diversity and selective regulatory mechanisms to its role in the control of cellular Ca2+ homeostasis.
Collapse
Affiliation(s)
- María Del Rocío Cantero
- Laboratorio de Canales Iónicos, Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD, CONICET-UNSE), El Zanjón, Santiago del Estero 4206, Argentina.
| | - Horacio F Cantiello
- Laboratorio de Canales Iónicos, Instituto Multidisciplinario de Salud, Tecnología y Desarrollo (IMSaTeD, CONICET-UNSE), El Zanjón, Santiago del Estero 4206, Argentina
| |
Collapse
|
5
|
Evidence of retinal arteriolar narrowing in patients with autosomal-dominant polycystic kidney disease. POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2022-0043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Abstract
Introduction
The aim of this study was to examine retinal vessels in autosomal dominant polycystic kidney disease (ADPKD) patients with normal kidney function and without diabetes mellitus.
Materials and Methods
We enrolled 39 adult individuals with ADPKD and 45 gender- and age-matched individuals as controls. A full ophthalmologic examination, including retinal vessel caliber and reactions to flicker stimulation analysis and grading of hypertensive retinopathy according to the Keith-Wagener classification, was performed.
Results
Multivariable analysis of ADPKD patients and controls, adjusted for age, gender, estimated glomerular filtration rate (e-GFR) and the presence of hypertension, revealed that ADPKD was an independent factor associated with lower arteriovenous ratio (AVR) values (by 0.069 on average, β = −0.50, p < 0.0001). The severity of hypertensive retinopathy according to the Keith-Wagener classification appeared to be more advanced in the ADPKD group than in the controls, despite the lack of vascular abnormalities, such as retinal hemorrhages, exudates, cotton wool spots or papilledema, as well as microaneurysms, which are very characteristic signs of ADPKD in other vascular beds.
Conclusions
Lower AVR values could be a specific pathophysiological ocular manifestation of systemic vasculopathy in the course of ADPKD.
Collapse
|
6
|
Evidence of retinal arteriolar narrowing in patients with autosomal-dominant polycystic kidney disease. POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2022-0001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Introduction
The aim of this study was to examine retinal vessels in autosomal dominant polycystic kidney disease (ADPKD) patients with normal kidney function and without diabetes mellitus.
Materials and Methods
We enrolled 39 adult individuals with ADPKD and 45 gender- and age-matched individuals as controls. A full ophthalmologic examination, including retinal vessel caliber and reactions to flicker stimulation analysis and grading of hypertensive retinopathy according to the Keith-Wagener classification, was performed.
Results
Multivariable analysis of ADPKD patients and controls, adjusted for age, gender, estimated glomerular filtration rate (e-GFR) and the presence of hypertension, revealed that ADPKD was an independent factor associated with lower arteriovenous ratio (AVR) values (by 0.069 on average, β = −0.50, p < 0.0001). The severity of hypertensive retinopathy according to the Keith-Wagener classification appeared to be more advanced in the ADPKD group than in the controls, despite the lack of vascular abnormalities, such as retinal hemorrhages, exudates, cotton wool spots or papilledema, as well as microaneurysms, which are very characteristic signs of ADPKD in other vascular beds.
Conclusions
Lower AVR values could be a specific pathophysiological ocular manifestation of systemic vasculopathy in the course of ADPKD.
Collapse
|
7
|
Localized cystic disease of the kidney in pediatric patients: Clinical and imaging findings with long term follow up. J Pediatr Urol 2022; 18:90.e1-90.e8. [PMID: 34862129 DOI: 10.1016/j.jpurol.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/07/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION This study aimed to describe the clinical and imaging findings, including ultrasonography (US) findings, and long-term follow-up results in pediatric patients with localized cystic disease of the kidney (LCDK). MATERIAL AND METHODS Retrospective review of pediatric patients diagnosed with LCDK based on imaging findings showing multiple localized renal cysts with intervening normal renal parenchyma from January 2002 to August 2020. Clinical presentations and US features of the affected and contralateral kidneys were reviewed and compared with computed tomography or magnetic resonance imaging findings, if available. RESULTS A total of 18 patients (male:female = 11:7; median age, 8 years) were included. Initial clinical presentations were incidental findings (n = 5), abdominal pain (n = 5), or hematuria (n = 5). Of the seven patients (7/18, 39%) who showed multiple hyperechoic foci with ring-down artifacts within the cystic lesions on US, six patients showed focal calcification of the cysts on CT. Two patients (2/18, 11%) had milimetric cysts in the contralateral kidney. During follow-up (range, 2-122 months), there was an increase in lesion size, with ipsilateral renal growth in four patients (4/18, 22%). There was no renal function impairment at the initial presentation or during follow-up in all patients. CONCLUSIONS LCDK can present with milimetric calcifications (39%), contralateral milimetric renal cysts (11%), and mild size increase during renal growth (22%) in children without renal function impairment. Follow-up US is recommended without surgical intervention in these typical cases.
Collapse
|
8
|
Murakami M, Murakami AM, Nemoto T, Ohba T, Yonekura M, Toyama Y, Tomita H, Matsuzaki Y, Sawamura D, Hirota K, Itagaki S, Asada Y, Miyoshi I. Enhanced β-adrenergic response in mice with dominant-negative expression of the PKD2L1 channel. PLoS One 2022; 17:e0261668. [PMID: 35051185 PMCID: PMC8775249 DOI: 10.1371/journal.pone.0261668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 12/08/2021] [Indexed: 11/19/2022] Open
Abstract
Polycystic kidney disease (PKD) is the most common genetic cause of kidney failure in humans. Among the various PKD-related molecules, PKD2L1 forms cation channels, but its physiological importance is obscure. In the present study, we established a transgenic mouse line by overexpressing the dominant-negative form of the mouse PKD2L1 gene (i.e., lacking the pore-forming domain). The resulting PKD2L1del-Tg mice exhibited supraventricular premature contraction, as well as enhanced sensitivity to β-adrenergic stimulation and unstable R-R intervals in electrocardiography. During spontaneous atrial contraction, PKD2L1del-Tg atria showed enhanced sensitivity to isoproterenol, norepinephrine, and epinephrine. Action potential recording revealed a shortened action potential duration in PKD2L1del-Tg atria in response to isoproterenol. These findings indicated increased adrenergic sensitivity in PKD2L1del-Tg mice, suggesting that PKD2L1 is involved in sympathetic regulation.
Collapse
Affiliation(s)
- Manabu Murakami
- Department of Pharmacology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Agnieszka M. Murakami
- Department of Pharmacology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Takayuki Nemoto
- Department of Pharmacology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Takayoshi Ohba
- Department of Cell Physiology, Akita University School of Medicine, Akita, Akita, Japan
| | - Manabu Yonekura
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yuichi Toyama
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Hirofumi Tomita
- Department of Cardiology and Nephrology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Yasushi Matsuzaki
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Daisuke Sawamura
- Department of Dermatology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Kazuyoshi Hirota
- Department of Anesthesiology, Hirosaki University Graduate School of Medicine, Hirosaki, Aomori, Japan
| | - Shirou Itagaki
- Collaboration Center for Community and Industry, Sapporo Medical University, Sapporo, Hokkaido, Japan
| | - Yujiro Asada
- Division of Pathophysiology, Department of Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki, Miyazaki, Japan
| | - Ichiro Miyoshi
- Department of Laboratory Animal Medicine, Tohoku University School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
9
|
Schirrer L, Marín-García PJ, Llobat L. Feline Polycystic Kidney Disease: An Update. Vet Sci 2021; 8:269. [PMID: 34822642 PMCID: PMC8625840 DOI: 10.3390/vetsci8110269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 11/16/2022] Open
Abstract
Polycystic kidney disease (PKD) is a disease that affects felines and other mammals, such as humans. The common name is autosomal dominant polycystic kidney disease (ADPKD) and causes a progressive development of fluid-filled cysts in the kidney and sometimes in other organs as the liver and pancreas. The formation and growth of cysts progress slowly, causing deterioration of kidney tissue and a gradual decrease in kidney function, leading to irreversible kidney failure. Feline PKD or ADPKD in humans are hereditary pathologies of autosomal dominant transmission. ADPKD is one of the genetic diseases with the highest prevalence in humans. In cats, this disease also has a high prevalence, mainly in the Persian breed, being one of the most common feline genetic diseases. Imaging tests seem to be the most reliable method for diagnosis of the disease, although more genetic tests are being developed to detect the presence of the responsible mutation. In this review, we summarize the current knowledge about feline PKD to guide future research related to an adequate diagnosis and early detection of causal mutations. It can allow the establishment of selection programs to reduce or eliminate this pathology in feline breeds.
Collapse
Affiliation(s)
| | - Pablo Jesús Marín-García
- Department of Animal Production and Health, Veterinary Public Health and Food Sciences and Technology (PASAPTA), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Valencia, Spain;
| | - Lola Llobat
- Department of Animal Production and Health, Veterinary Public Health and Food Sciences and Technology (PASAPTA), Facultad de Veterinaria, Universidad Cardenal Herrera-CEU, CEU Universities, 46113 Valencia, Spain;
| |
Collapse
|
10
|
Pietrzak-Nowacka M, Safranow K, Czechowska M, Dutkiewicz G, Gątarska E, Ciechanowski K. Cardiac involvement in patients with Autosomal
Dominant Polycystic Kidney Disease and normal
renal function after six years of follow-up. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0015.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The aim of the follow-up study was to compare the changes of M-mode echocardiographic
parameters in autosomal dominant polycystic kidney disease (ADPKD) patients and controls
without renal failure during six years of observation and to explore the associations of these
parameters with metabolic syndrome components and kidney function. We performed
a follow-up examination in 37 ADPKD patients and 40 controls. Anthropometric parameters
were measured and fasting venous blood sample from each patient was tested for glucose,
insulin, C-peptide, HbA1c, creatinine, and urea concentrations. All subjects underwent standard
two-dimensional M-mode echocardiography. Left ventricular hypertrophy (LVH) was
diagnosed based on left ventricular mass index (LVMI) adjusted for body surface area (LVMI-
-S, LVH-S) or for height (LVMI-H, LVH-H). The prevalence of LVH was significantly greater
in ADPKD patients than in controls (35% vs. 10%, p=0.012) according to the ESH/ESC criteria
from 2013, and (27.0% vs. 7.5%, p=0.032) according to criteria from 2017. In patients with
ADPKD, no significant increase of the echocardiographic parameters was observed in the
6 years between the initial examination and the follow-up examination. Cardiac involvement
in women with ADPKD occurs at an earlier stage of the disease than in men. In patients with
ADPKD treated for hypertension in accordance with the 2013 ESH/ESC Guidelines the progression
of LVH was not observed during the 6-year follow-up, despite the deterioration of
renal function. Obesity, blood pressure and renal function do not affect LVMI changes.
Collapse
Affiliation(s)
- Maria Pietrzak-Nowacka
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | | | - Grażyna Dutkiewicz
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Ewa Gątarska
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Kazimierz Ciechanowski
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
11
|
Brill AL, Ehrlich BE. Polycystin 2: A calcium channel, channel partner, and regulator of calcium homeostasis in ADPKD. Cell Signal 2019; 66:109490. [PMID: 31805375 DOI: 10.1016/j.cellsig.2019.109490] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/30/2019] [Accepted: 12/01/2019] [Indexed: 01/26/2023]
Abstract
Polycystin 2 (PC2) is one of two main protein types responsible for the underlying etiology of autosomal dominant polycystic kidney disease (ADPKD), the most prevalent monogenic renal disease in the world. This debilitating and currently incurable condition is caused by loss-of-function mutations in PKD2 and PKD1, the genes encoding for PC2 and Polycystin 1 (PC1), respectively. Two-hit mutation events in these genes lead to renal cyst formation and eventual kidney failure, the main hallmarks of ADPKD. Though much is known concerning the physiological consequences and dysfunctional signaling mechanisms resulting from ADPKD development, to best understand the requirement of PC2 in maintaining organ homeostasis, it is important to recognize how PC2 acts under normal conditions. As such, an array of work has been performed characterizing the endogenous function of PC2, revealing it to be a member of the transient receptor potential (TRP) channel family of proteins. As a TRP protein, PC2 is a nonselective, cation-permeant, calcium-sensitive channel expressed in all tissue types, where it localizes primarily on the endoplasmic reticulum (ER), primary cilia, and plasma membrane. In addition to its channel function, PC2 interacts with and acts as a regulator of a number of other channels, ultimately further affecting intracellular signaling and leading to dysfunction in its absence. In this review, we describe the biophysical and physiological properties of PC2 as a cation channel and modulator of intracellular calcium channels, along with how these properties are altered in ADPKD.
Collapse
Affiliation(s)
- Allison L Brill
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA; Department of Pharmacology, Yale University, New Haven, CT, USA.
| |
Collapse
|
12
|
Pietrzak-Nowacka M, Safranow K, Marchelek-Myśliwiec M, Bodnar M, Przysiecka S, Nowosiad-Magda M, Ciechanowski K. Evolution of metabolic syndrome components in patients with autosomal-dominant polycystic kidney disease: a six-year follow-up study. POSTEP HIG MED DOSW 2019. [DOI: 10.5604/01.3001.0013.5605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Aim: Long-term studies show that some metabolic syndrome (MS) components deteriorate renal function in autosomal dominant polycystic kidney disease (ADPKD) patients. The aim of this 6-year follow-up was to analyze early changes of all MS components and their associations with kidney function in the nondiabetic ADPKD patients with normal renal function, compared to controls. Material/Methods: The follow-up physical and laboratory examinations were performed for 39 ADPKD patients (age 43.7 ± 11.4 years) and 44 controls (43.5 ± 9.1 years). Results: We noticed a significant increase in weight, body mass index (BMI), waist, total and LDL cholesterol, C-peptide, uric acid, creatinine and significant decline of HbA1c and e-GFR in the ADPKD group. Increases in waist, uric acid and creatinine concentrations were significantly higher in the ADPKD patients than controls. Both groups showed similar rates of prediabetes, while diabetes developed in 5 controls (with 4 cases of type 2 diabetes and one case of type 1), but not in the ADPKD group (11% vs 0%, P = 0.06 for diabetes, 9% vs 0%, P = 0.12 for type 2 diabetes). The ADPKD group showed a significantly higher percentage of obesity, waist circumferences, systolic/diastolic blood pressure, concentrations of creatinine, urea and uric acid and lower e-GFR. The MS prevalence was comparable; however, the number of MS components was significantly higher in the ADPKD patients (median 2 vs. 1, p = 0.001). Conclusions: The presence of MS does not influence the rate of renal failure progression in nondiabetic ADPKD patients with normal renal function at a 6-year follow-up.
Collapse
Affiliation(s)
- Maria Pietrzak-Nowacka
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Krzysztof Safranow
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Szczecin, Poland
| | | | - Mariusz Bodnar
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Sylwia Przysiecka
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Monika Nowosiad-Magda
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | - Kazimierz Ciechanowski
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
13
|
Polycystin-1 Inhibits Cell Proliferation through Phosphatase PP2A/B56 α. BIOMED RESEARCH INTERNATIONAL 2019; 2019:2582401. [PMID: 31641668 PMCID: PMC6770331 DOI: 10.1155/2019/2582401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/21/2019] [Accepted: 08/21/2019] [Indexed: 01/15/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is associated with a number of cellular defects such as hyperproliferation, apoptosis, and dedifferentiation. Mutations in polycystin-1 (PC1) account for ∼85% of ADPKD. Here, we showed that wild-type (WT) or mutant PC1 composed of the last five transmembrane (TM) domains and the C-terminus (termed PC1-5TMC) inhibits cell proliferation and protein translation, as well as the downstream effectors of mTOR, consistent with previous reports. Knockdown of B56α, a subunit of the protein phosphatase 2A (PP2A) complex, or application of PP2A inhibitor okadaic acid or calyculin A, abolished the inhibitory effect of PC1 and PC1-5TMC on proliferation, indicating that PP2A/B56α mediates the regulation of cell proliferation by PC1. In addition to the phosphorylated S6 and 4EBP1, B56α was also downregulated by PC1 and PC1-5TMC. Furthermore, the downregulation of B56α, which may be mediated by mTOR but not AKT, can account for the dependence of PC1-inhibited proliferation on PP2A.
Collapse
|
14
|
Role of PKR in the Inhibition of Proliferation and Translation by Polycystin-1. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5320747. [PMID: 31341901 PMCID: PMC6612395 DOI: 10.1155/2019/5320747] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/19/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is mainly caused by mutations in the PKD1 (~85%) or PKD2 (~15%) gene which, respectively, encode polycystin-1 (PC1) and polycystin-2 (PC2). How PC1 regulates cell proliferation and apoptosis has been studied for decades but the underlying mechanisms remain controversial. Protein kinase RNA-activated (PKR) is activated by interferons or double-stranded RNAs, inhibits protein translation, and induces cell apoptosis. In a previous study, we found that PC1 reduces apoptosis through suppressing the PKR/eIF2α signaling. Whether and how PKR is involved in PC1-inhibited proliferation and protein synthesis remains unknown. Here we found that knockdown of PKR abolishes PC1-inhibited proliferation and translation. Because suppressed PKR-eIF2α signaling/activity by PC1 would stimulate, rather than inhibit, the proliferation and translation, we examined the effect of dominant negative PKR mutant K296R that has no kinase activity and found that it enhances the inhibition of proliferation and translation by PC1. Thus, our study showed that inhibition of cell proliferation and protein synthesis by PC1 is mediated by the total expression but not the kinase activity of PKR, possibly through physical association.
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This review provides an outline of the most recent insights and significant discoveries regarding the genetic mechanisms involved in polycystic liver disease. RECENT FINDINGS Polycystic liver disease includes a heterogeneous group of genetic disorders characterized by multiple hepatic cysts. Isolated liver cysts are caused by mutations in Protein Kinase C Substrate 80K-H (PRKCSH), SEC63, and LDL Receptor Related Protein 5 (LRP5), whereas Polycystic Kidney Disease (PKD)1, PKD2, and PKHD1 mutations cause kidney cysts often accompanied by liver cysts. Glucosidase II Alpha Subunit (GANAB) has been reported to cause both phenotypes. These mutations, together with the newly identified ones in SEC61B and Alpha-1,3-Glucosyltransferase (ALG8), can be found in ∼50% of patients with isolated polycystic liver disease. Somatic second hit-mutations are hypothesized as driving force leading to cystogenesis. Subsequently, loss of heterozygosity in the cystic tissue aggravates disease progression. All genetic mutations lead to reduced levels of functional polycystin-1. This ciliary protein is therefore considered to be the central factor in the development and severity of liver cysts. SUMMARY Recent advances of the genetic complexity leading to hepatic cystogenesis provide novel candidate genes and important mechanistic insights with polycystin-1 as a common denominator.
Collapse
|
16
|
He WB, Xiao WJ, Tan YQ, Zhao XM, Li W, Zhang QJ, Zhong CG, Li XR, Hu L, Lu GX, Lin G, Du J. Novel mutations of PKD genes in Chinese patients suffering from autosomal dominant polycystic kidney disease and seeking assisted reproduction. BMC MEDICAL GENETICS 2018; 19:186. [PMID: 30333007 PMCID: PMC6192368 DOI: 10.1186/s12881-018-0693-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/24/2018] [Indexed: 01/24/2023]
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD), the commonest inherited kidney disease, is generally caused by heterozygous mutations in PKD1, PKD2, or GANAB (PKD3). Methods We performed mutational analyses of PKD genes to identify causative mutations. A set of 90 unrelated families with ADPKD were subjected to mutational analyses of PKD genes. Genes were analysed using long-range PCR (LR-PCR), direct PCR sequencing, followed by multiplex ligation-dependent probe amplification (MLPA) or screening of GANAB for some patients. Semen quality was assessed for 46 male patients, and the correlation between mutations and male infertility was analysed. Results A total of 76 mutations, including 38 novel mutations, were identified in 77 families, comprising 72 mutations in PKD1 and 4 in PKD2, with a positive detection rate of 85.6%. No pathogenic mutations of GANAB were detected. Thirty-seven patients had low semen quality and were likely to be infertile. No association was detected between PKD1 mutation type and semen quality. However, male patients carrying a pathogenic mutation in the Ig-like repeat domain of PKD1 had a high risk of infertility. Conclusion Our study identified a group of novel mutations in PKD genes, which enrich the PKD mutation spectrum and might help clinicians to make precise diagnoses, thereby allowing better family planning and genetic counselling. Men with ADPKD accompanied by infertility should consider intracytoplasmic sperm injection combined with preimplantation genetic diagnosis to achieve paternity and obtain healthy progeny. Electronic supplementary material The online version of this article (10.1186/s12881-018-0693-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Wen-Bin He
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Wen-Juan Xiao
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China
| | - Yue-Qiu Tan
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Xiao-Meng Zhao
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Wen Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Qian-Jun Zhang
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Chang-Gao Zhong
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Xiu-Rong Li
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Liang Hu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Guang-Xiu Lu
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Ge Lin
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China.,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China
| | - Juan Du
- Institute of Reproductive and Stem Cell Engineering, School of Basic Medical Science, Central South University, Changsha, Hunan, 410078, People's Republic of China. .,Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, Hunan, 410078, People's Republic of China.
| |
Collapse
|
17
|
Elisakova V, Merta M, Reiterova J, Baxova A, Kotlas J, Hirschfeldova K, Obeidova L, Tesar V, Stekrova J. Bilineal inheritance of pathogenic PKD1 and PKD2 variants in a Czech family with autosomal dominant polycystic kidney disease - a case report. BMC Nephrol 2018; 19:163. [PMID: 29973168 PMCID: PMC6032778 DOI: 10.1186/s12882-018-0978-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 06/28/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary renal disorder, leading to end stage renal failure and kidney transplantation in its most serious form. The severity of the disease's manifestation depends on the genetic determination of ADPKD. The huge variability of different phenotypes (even within a single family) is not only modulated by the two main ADPKD genes (PKD1 and PKD2) but also by modifier genes and the whole genetic background. CASE PRESENTATION This is a report of an ADPKD family with co-inheritance of PKD1 and PKD2 pathogenic variants. The proband, with an extremely serious manifestation of ADPKD (the man was diagnosed in early childhood, and with end stage renal disease aged 23), underwent genetic analysis of PKD1 and PKD2, which revealed the presence of pathogenic mutations in both of these genes. The missense PKD2 mutation p.Arg420Gly came from the proband's father, with a mild ADPKD phenotype. The same mutation of the PKD2 gene and similar mild disease presentation were found in the proband's aunt (father's sister) and her son. The nonsense mutation p.Gln2196* within the PKD1 gene was probably inherited from the proband's mother, who died at the age of 45. It was only discovered post mortem, that the real cause of her death was kidney failure as a consequence of untreated ADPKD. Unfortunately, neither the DNA of the proband's mother nor the DNA of any other family members from this side of the pedigree were available for further examination. The proband underwent successful cadaveric kidney transplantation at the age of 24, and this replacement therapy lasted for the next 15 years. CONCLUSIONS Here, we present a first case of bilineal ADPKD inheritance in the Czech Republic. This report highlights the significant role of modifier genes in genetic determination of ADPKD, especially in connection with seriously deteriorated disease phenotypes. In our case, the modifying role is probably mediated by the PKD2 gene.
Collapse
Affiliation(s)
- Veronika Elisakova
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Miroslav Merta
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Jana Reiterova
- Department of Nephrology, First Faculty of Medicine Charles University and General University Hospital in Prague, U Nemocnice 2, 128 00, Prague, Czech Republic
| | - Alica Baxova
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Jaroslav Kotlas
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Katerina Hirschfeldova
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Lena Obeidova
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic
| | - Vladimir Tesar
- Department of Nephrology, First Faculty of Medicine Charles University and General University Hospital in Prague, U Nemocnice 2, 128 00, Prague, Czech Republic
| | - Jitka Stekrova
- Institute of Biology and Medical Genetics, First Faculty of Medicine Charles University and General University Hospital in Prague, Albertov 4, 128 00, Prague, Czech Republic.
| |
Collapse
|
18
|
Kocyigit I, Eroglu E, Gungor O. Clinical problems in hemodialysis patients with autosomal dominant polycystic kidney disease. Semin Dial 2018; 31:268-277. [DOI: 10.1111/sdi.12696] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Ismail Kocyigit
- Department of Nephrology; Erciyes University Medical Faculty; Kayseri Turkey
| | - Eray Eroglu
- Department of Nephrology; Erciyes University Medical Faculty; Kayseri Turkey
| | - Ozkan Gungor
- Department of Nephrology; Sutcu Imam University Medical Faculty; Kahramanmaras Turkey
| |
Collapse
|
19
|
Genetic diagnosis of polycystic kidney disease, Alport syndrome, and thalassemia minor in a large Chinese family. Clin Sci (Lond) 2017; 131:2427-2438. [PMID: 28827396 DOI: 10.1042/cs20170245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 07/31/2017] [Accepted: 08/16/2017] [Indexed: 01/13/2023]
Abstract
Polycystic kidney disease (PKD) and Alport syndrome (AS) are serious inherited disorders associated with renal disease, and thalassemia is a hereditary blood disease with a high prevalence in south China. Here, we report an exceptional PKD coincidence of thalassemia minor and AS (diagnosed genetically) in a large Chinese family. Whole genome next-generation sequencing (NGS) was performed on the proband, and all family members underwent clinical evaluation. Sanger sequencing was used to validate the mutations distinguished by NGS. The pathogenic potential of the variants were evaluated by Polymorphism Phenotyping v2 (PolyPhen-2), Sorting Intolerant From Tolerant (SIFT) algorithm, and MutationTaster. Immunohistochemical, Western blot, immunofluorescent, and TdT-mediated dUTP nick-end labeling (TUNEL) analyses were performed to investigate polycystin 1 (PC1) expression, and cell proliferation and apoptosis in kidney tissues from the proband and normal control. A novel frameshift polycystic kidney disease 1 (PKD1) mutation (c.3903delC, p.A1302Pfs) was identified to be responsible for renal disease in this family. PC1 expression, and cell proliferation and apoptosis were significantly increased in the kidney tissues of the proband. Moreover, a deletion of approximately 19.3 kb of DNA with α-globin genes (_ _SEA) was associated with thalassemia minor in the family. In addition, a collagen type IV α 5 chain (COL4A5) variant (c.2858G>T, rs78972735), annotated as a pathogenic mutation in dbSNP and human gene mutation database (HGMD), was found in four family members with no clinical traits of AS. A novel pathogenic PKD1 mutation (c.3903delC) and (_ _SEA) thalassemia deletion were found to be responsible for the clinical symptoms in this family. The reported pathogenic COL4a5 variant (c.2858G>T, rs78972735) was not pathogenic alone.
Collapse
|
20
|
Fang Z, Xu S, Wang Y, Sun L, Feng Y, Guo Y, Li H, Jiang W. Pathogenicity analysis of novel variations in Chinese Han patients with polycystic kidney disease. Gene 2017; 626:433-441. [PMID: 28578020 DOI: 10.1016/j.gene.2017.05.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/24/2017] [Accepted: 05/22/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Locus and allellic heterogeneity in polycystic kidney disease (PKD) is a great challenge in precision diagnosis. We aim to establish comprehensive methods to distinguish the pathogenic mutations from the variations in PKD1, PKD2 and PKHD1 genes in a limited time and lay the foundation for precisely prenatal diagnosis, preimplantation genetic diagnosis and presymptom diagnosis of PKD. METHODS Nested PCR combined with direct DNA sequencing were used to screen variations in PKD1, PKD2 and PKHD1 genes. The pathogenicity of de novel variations was assessed by the comprehensive methods including clinic data and literature review, databases query, analysis of co-segregation of the variants with the disease, variant frequency screening in the population, evolution conservation comparison, protein structure analysis and splice sites predictions. RESULTS 17 novel mutations from 15 Chinese Han families were clarified including 10 mutations in PKD1 gene and 7 mutations in PKHD1 gene. The novel mutations were classified as 4 definite pathogenic, 2 highly likely pathogenic, 4 likely pathogenic, 7 indeterminate by the comprehensive analysis. The results were verified the truth by the follow-up visits. CONCLUSIONS The comprehensive methods may be useful in distinguishing the pathogenic mutations from the variations in PKD1, PKD2 and PKHD1 genes for prenatal diagnosis and presymptom diagnosis of PKD. Our results also enriched PKD genes mutation spectrum and evolved possible genotype-phenotype correlations of Chinese Han population.
Collapse
Affiliation(s)
- Zishui Fang
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Shiyan Xu
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; ShenZhen People's Hospital, China
| | - Yonghua Wang
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Liwei Sun
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Feng
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yibin Guo
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Hongyi Li
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Weiying Jiang
- Department of Medical Genetics, ZhongShan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
21
|
The mutation-free embryo for in vitro fertilization selected by MALBAC-PGD resulted in a healthy live birth from a family carrying PKD 1 mutation. J Assist Reprod Genet 2017; 34:1653-1658. [PMID: 28825164 DOI: 10.1007/s10815-017-1018-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Accepted: 08/02/2017] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD, autosomal dominant PKD or adult-onset PKD) is the most prevalent and potentially lethal kidney disease that is hereditary and lacks effective treatment. Preimplantation genetic diagnosis (PGD) of embryos in assistant reproductive technology (ART) helps to select mutation-free embryos for blocking ADPKD inheritance from the parents to their offspring. However, there are multiple pseudogenes in the PKD1 coding region, which make blocking ADPKD inheritance by PGD complicated and difficult. Therefore, this technique has not been recommended and used routinely to ADPKD family plan. METHODS AND RESULTS Here, we report a new strategy of performing PGD in screening (target-) mutation-free embryos. We firstly used a long-range PCR amplification and next generation sequencing to identify the potential PKD1 mutant(s). After pathogenic variants were detected, multiple annealing and looping-based amplification cycles (MALBAC), a recently developed whole genome amplification method, was used to screen embryo cells. We successfully distinguished the mutated allele among pseudogenes and obtained mutation-free embryos for implantation. The first embryo transfer attempt resulted in a healthy live birth free of ADPKD condition and chromosomal anomalies which was confirmed by aminocentesis at week 18 of gestation, and by performing live birth genetic screening. CONCLUSIONS The first MALBAC-PGD attempt in ADPKD patient resulted in a healthy live birth free of ADPKD and chromosomal anomalies. MALBAC-PGD also enables selecting embryos without aneuploidy together and target gene mutation, thereby increasing implantation and live birth rates.
Collapse
|
22
|
Genetically modified pigs to model human diseases. J Appl Genet 2015; 55:53-64. [PMID: 24234401 DOI: 10.1007/s13353-013-0182-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 10/22/2013] [Indexed: 01/06/2023]
Abstract
Genetically modified mice are powerful tools to investigate the molecular basis of many human diseases. Mice are, however, of limited value for preclinical studies, because they differ significantly from humans in size, general physiology, anatomy and lifespan. Considerable efforts are, thus, being made to develop alternative animal models for a range of human diseases. These promise powerful new resources that will aid the development of new diagnostics, medicines and medical procedures. Here, we provide a comprehensive review of genetically modified porcine models described in the scientific literature: various cancers, cystic fibrosis, Duchenne muscular dystrophy, autosomal polycystic kidney disease, Huntington’s disease, spinal muscular atrophy, haemophilia A, X-linked severe combined immunodeficiency, retinitis pigmentosa, Stargardt disease, Alzheimer’s disease, various forms of diabetes mellitus and cardiovascular diseases.
Collapse
|
23
|
He J, Li Q, Fang S, Guo Y, Liu T, Ye J, Yu Z, Zhang R, Zhao Y, Hu X, Bai X, Chen X, Li N. PKD1 mono-allelic knockout is sufficient to trigger renal cystogenesis in a mini-pig model. Int J Biol Sci 2015; 11:361-9. [PMID: 25798056 PMCID: PMC4366635 DOI: 10.7150/ijbs.10858] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 12/03/2014] [Indexed: 11/13/2022] Open
Abstract
PKD1 and PKD2 mutations could lead to autosomal dominant polycystic kidney disease (ADPKD), which afflicts millions of people worldwide. Due to the marked differences in the lifespan, size, anatomy, and physiology from humans, rodent ADPKD models cannot fully mimic the disease. To obtain a large animal model that recapitulates the disease, we constructed a mini-pig model by mono-allelic knockout (KO) of PKD1 using zinc finger nuclease. The mono-allelic KO pigs had lower PKD1 expression than their wild-type littermates at both the transcriptional and translational levels. After approximately six months, renal cysts appeared and grew progressively in the KO pigs. Histological analysis showed that renal cysts were scatteredly distributed in the mutant pig kidneys and were lined by either cuboidal or flattened epithelial cells. Contrast-enhanced computed tomography confirmed that all of the mutant pigs had renal and hepatic cysts, when they were 11-month-old. Immunohistochemical analysis revealed that most of the cysts were derived from the proximal tubules and collecting ducts. Therefore, the PKD1 mono-allelic knockout is sufficient to trigger renal cystogenesis, and this pig model may provide a platform for future study of renal cyst formation.
Collapse
Affiliation(s)
- Jin He
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China ; 2. College of Animal Science and Technology, China Agricultural University, Beijing, PR China
| | - Qiuyan Li
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Suyun Fang
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Ying Guo
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Tongxin Liu
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Jianhua Ye
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Zhengquan Yu
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Ran Zhang
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Yaofeng Zhao
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Xiaoxiang Hu
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China
| | - Xueyuan Bai
- 3. Department of Nephrology, State Key Laboratory of Kidney Disease, Chinese PLA General Hospital, Beijing, PR China
| | - Xiangmei Chen
- 3. Department of Nephrology, State Key Laboratory of Kidney Disease, Chinese PLA General Hospital, Beijing, PR China
| | - Ning Li
- 1. State Key Laboratory for Agrobiotechnology, College of Biological Sciences, China Agricultural University, Beijing, PR China ; 4. College of Animal Science and Technology, Yunnan Agricultural University, Kunming, PR China
| |
Collapse
|
24
|
Novel mutations of PKD genes in the Czech population with autosomal dominant polycystic kidney disease. BMC MEDICAL GENETICS 2014; 15:41. [PMID: 24694054 PMCID: PMC3992149 DOI: 10.1186/1471-2350-15-41] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 03/10/2014] [Indexed: 11/10/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary renal disorder caused by mutation in either one of two genes, PKD1 and PKD2. High structural and sequence complexity of PKD genes makes the mutational diagnostics of ADPKD challenging. The present study is the first detailed analysis of both PKD genes in a cohort of Czech patients with ADPKD using High Resolution Melting analysis (HRM) and Multiplex Ligation-dependent Probe Amplification (MLPA). METHODS The mutational analysis of PKD genes was performed in a set of 56 unrelated patients. For mutational screening of the PKD1 gene, the long-range PCR (LR-PCR) strategy followed by nested PCR was used. Resulting PCR fragments were analyzed by HRM; the positive cases were reanalyzed and confirmed by direct sequencing. Negative samples were further examined for sequence changes in the PKD2 gene by the method of HRM and for large rearrangements of both PKD1 and PKD2 genes by MLPA. RESULTS Screening of the PKD1 gene revealed 36 different likely pathogenic germline sequence changes in 37 unrelated families/individuals. Twenty-five of these sequence changes were described for the first time. Moreover, a novel large deletion was found within the PKD1 gene in one patient. Via the mutational analysis of the PKD2 gene, two additional likely pathogenic mutations were detected. CONCLUSIONS Probable pathogenic mutation was detected in 71% of screened patients. Determination of PKD mutations and their type and localization within corresponding genes could help to assess clinical prognosis of ADPKD patients and has major benefit for prenatal and/or presymptomatic or preimplantational diagnostics in affected families as well.
Collapse
|
25
|
Renal expression of FGF23 and peripheral resistance to elevated FGF23 in rodent models of polycystic kidney disease. Kidney Int 2014; 85:1340-50. [PMID: 24402093 DOI: 10.1038/ki.2013.526] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Revised: 09/11/2013] [Accepted: 10/17/2013] [Indexed: 11/08/2022]
Abstract
Fibroblast growth factor 23 (FGF23) regulates phosphate homeostasis and is linked to cardiovascular disease and all-cause mortality in chronic kidney disease. FGF23 rises in patients with CKD stages 2-3, but in patients with autosomal dominant polycystic kidney disease, the increase of FGF23 precedes the first measurable decline in renal function. The mechanisms governing FGF23 production and effects in kidney disease are largely unknown. Here we studied the relation between FGF23 and mineral homeostasis in two animal models of PKD. Plasma FGF23 levels were increased 10-fold in 4-week-old cy/+ Han:SPRD rats, whereas plasma urea and creatinine concentrations were similar to controls. Plasma calcium and phosphate levels as well as TmP/GFR were similar in PKD and control rats at all time points examined. Expression and activity of renal phosphate transporters, the vitamin D3-metabolizing enzymes, and the FGF23 co-ligand Klotho in the kidney were similar in PKD and control rats through 8 weeks of age, indicating resistance to FGF23, although phosphorylation of the FGF receptor substrate 2α protein was enhanced. In the kidneys of rats with PKD, FGF23 mRNA was highly expressed and FGF23 protein was detected in cells lining renal cysts. FGF23 expression in bone and spleen was similar in control rats and rats with PKD. Similarly, in an inducible Pkd1 knockout mouse model, plasma FGF23 levels were elevated, FGF23 was expressed in kidneys, but renal phosphate excretion was normal. Thus, the polycystic kidney produces FGF23 but is resistant to its action.
Collapse
|
26
|
Evidence of a third ADPKD locus is not supported by re-analysis of designated PKD3 families. Kidney Int 2013; 85:383-92. [PMID: 23760289 PMCID: PMC3883953 DOI: 10.1038/ki.2013.227] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2013] [Revised: 03/29/2013] [Accepted: 05/02/2013] [Indexed: 01/18/2023]
Abstract
Mutations to PKD1 and PKD2 are associated with autosomal dominant polycystic kidney disease (ADPKD). The absence of apparent PKD1/PKD2 linkage in five published European or North American families with ADPKD suggested a third locus, designated PKD3. Here we re-evaluated these families by updating clinical information, re-sampling where possible, and mutation screening for PKD1/PKD2. In the French-Canadian family we identified PKD1: p.D3782_V3783insD, with misdiagnoses in two individuals and sample contamination explaining the lack of linkage. In the Portuguese family, PKD1: p.G3818A segregated with the disease in 10 individuals in three generations with likely misdiagnosis in one individual, sample contamination, and use of distant microsatellite markers explaining the linkage discrepancy. The mutation, PKD2: c.213delC, was found in the Bulgarian family, with linkage failure attributed to false positive diagnoses in two individuals. An affected son but not the mother, in the Italian family had the nonsense mutation, PKD1: p.R4228X, which appeared de novo in the son; with simple cysts probably explaining the mother’s phenotype. No likely mutation was found in the Spanish family, but the phenotype was atypical with kidney atrophy in one case. Thus, re-analysis does not support the existence of a PKD3 in ADPKD. False positive diagnoses by ultrasound in all resolved families shows the value of mutation screening, but not linkage, to understand families with discrepant data.
Collapse
|
27
|
Blanco G, Wallace DP. Novel role of ouabain as a cystogenic factor in autosomal dominant polycystic kidney disease. Am J Physiol Renal Physiol 2013; 305:F797-812. [PMID: 23761677 DOI: 10.1152/ajprenal.00248.2013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
The classic role of the Na-K-ATPase is that of a primary active transporter that utilizes cell energy to establish and maintain transmembrane Na(+) and K(+) gradients to preserve cell osmotic stability, support cell excitability, and drive secondary active transport. Recent studies have revealed that Na-K-ATPase located within cholesterol-containing lipid rafts serves as a receptor for cardiotonic steroids, including ouabain. Traditionally, ouabain was viewed as a toxin produced only in plants, and it was used in relatively high concentrations to experimentally block the pumping action of the Na-K-ATPase. However, the new and unexpected role of the Na-K-ATPase as a signal transducer revealed a novel facet for ouabain in the regulation of a myriad of cell functions, including cell proliferation, hypertrophy, apoptosis, mobility, and metabolism. The seminal discovery that ouabain is endogenously produced in mammals and circulates in plasma has fueled the interest in this endogenous molecule as a potentially important hormone in normal physiology and disease. In this article, we review the role of the Na-K-ATPase as an ion transporter in the kidney, the experimental evidence for ouabain as a circulating hormone, the function of the Na-K-ATPase as a signal transducer that mediates ouabain's effects, and novel results for ouabain-induced Na-K-ATPase signaling in cystogenesis of autosomal dominant polycystic kidney disease.
Collapse
Affiliation(s)
- Gustavo Blanco
- Dept. of Molecular and Integrative Physiology, 3901 Rainbow Blvd., Kansas City, KS 66160.
| | | |
Collapse
|
28
|
Construction of a transgenic pig model overexpressing polycystic kidney disease 2 (PKD2) gene. Transgenic Res 2013; 22:861-7. [PMID: 23315160 DOI: 10.1007/s11248-012-9686-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Accepted: 12/28/2012] [Indexed: 12/13/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common human genetic disease, affecting millions of people worldwide. The progressive growth of cysts in kidneys eventually leads to renal failure in 50 % of patients, and there is currently no effective treatment. Various murine models have been studied to elucidate the disease mechanisms, and much information has been acquired. However, the course of the disease cannot be fully recapitulated using these models. The pig is a suitable model for biomedical research, and pig PKD2 has high similarity to the human ortholog at the molecular level. Here, a mini-pig PKD2 transgenic model was generated, driven by a ubiquitous cytomegalovirus enhancer/promoter. Using somatic cell nuclear transfer, four transgenic pigs with approximately 10 insertion events each were generated. Quantitative real-time PCR and western blotting showed that PKD2 was more highly expressed in transgenic pigs than in wild-type counterparts. Because of the chronic nature of ADPKD, blood urea nitrogen and serum creatinine levels were continuously measured to assess the pig kidney function. The transgenic pigs continue to show no significant alteration in kidney function; it is estimated that 1-2 more years may be required for manifestation of renal cystogenesis in these pigs.
Collapse
|
29
|
Correas JM, Joly D, Chauveau D, Richard S, Hélénon O. Insuffisance rénale et maladies kystiques du rein. ACTA ACUST UNITED AC 2011; 92:308-22. [DOI: 10.1016/j.jradio.2011.02.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 02/25/2011] [Indexed: 11/24/2022]
|
30
|
Wang Q, Yin H, He J, Ye J, Ding F, Wang S, Hu X, Meng Q, Li N. cDNA cloning of porcine PKD2 gene and RNA interference in LLC-PK1 cells. Gene 2011; 476:38-45. [PMID: 21277361 DOI: 10.1016/j.gene.2011.01.017] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2010] [Revised: 01/11/2011] [Accepted: 01/24/2011] [Indexed: 11/16/2022]
Abstract
Mutations in the PKD2 gene cause autosomal dominant polycystic kidney disease (ADPKD), a common, inherited disease that frequently leads to end-stage renal disease (ESRD). Swine show substantial similarity to humans physiologically and anatomically, and are therefore a good model system in which to decipher the structure and function of the PKD2 gene and to identify potential therapeutic targets. Here we report the cloning and characterization of the porcine PKD2 cDNA showing that the full-length gene (3370 bases) is highly expressed in kidney, with minimal expression in the liver. RNA interference (RNAi) is a promising tool to enable identification of the essential components necessary for exploitation of the pathway involved in cellular processes. We therefore designed four shRNAs and nine siRNAs targeting the region of the porcine PKD2 gene from exons 3 to 9, which is supposed to be a critical region contributing to the severity of ADPKD. The results from HeLa cells with the dual-luciferase reporter system and porcine kidney cells (LLC-PK1) showed that sh12 could efficiently knock down the PKD2 gene with an efficiency of 51% and P1 and P2 were the most effective siRNAs inhibiting 85% and 77% respectively of PKD2 expression compared with untreated controls. A subsequent functional study of the transient receptor potential polycystic (TRPP) 2 channel protein indicated that the decreased expression of TRPP2 induced by siRNA P1 and P2 could release the arrest of the cell cycle from G0/G1 promoting progression to S and G2 phases. Our data, therefore, provides evidence of potential knock-down target sites in the PKD2 gene and paves the way for the future generation of transgenic ADPKD knock-down animal models.
Collapse
Affiliation(s)
- Qingsong Wang
- Tianjin Research Centre of Basic Medical Science, Tianjin Medical University, Qixiangtai Road, Heping District, Tianjin, 300070, China
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Boucher CA, Ward HH, Case RL, Thurston KS, Li X, Needham A, Romero E, Hyink D, Qamar S, Roitbak T, Powell S, Ward C, Wilson PD, Wandinger-Ness A, Sandford RN. Receptor protein tyrosine phosphatases are novel components of a polycystin complex. Biochim Biophys Acta Mol Basis Dis 2010; 1812:1225-38. [PMID: 21126580 DOI: 10.1016/j.bbadis.2010.11.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2010] [Revised: 11/16/2010] [Accepted: 11/19/2010] [Indexed: 12/27/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutation of PKD1 and PKD2 that encode polycystin-1 and polycystin-2. Polycystin-1 is tyrosine phosphorylated and modulates multiple signaling pathways including AP-1, and the identity of the phosphatases regulating polycystin-1 are previously uncharacterized. Here we identify members of the LAR protein tyrosine phosphatase (RPTP) superfamily as members of the polycystin-1complex mediated through extra- and intracellular interactions. The first extracellular PKD1 domain of polycystin-1 interacts with the first Ig domain of RPTPσ, while the polycystin-1 C-terminus of polycystin-1 interacts with the regulatory D2 phosphatase domain of RPTPγ. Additional homo- and heterotypic interactions between RPTPs recruit RPTPδ. The multimeric polycystin protein complex is found localised in cilia. RPTPσ and RPTPδ are also part of a polycystin-1/E-cadherin complex known to be important for early events in adherens junction stabilisation. The interaction between polycystin-1 and RPTPγ is disrupted in ADPKD cells, while RPTPσ and RPTPδ remain closely associated with E-cadherin, largely in an intracellular location. The polycystin-1 C-terminus is an in vitro substrate of RPTPγ, which dephosphorylates the c-Src phosphorylated Y4237 residue and activates AP1-mediated transcription. The data identify RPTPs as novel interacting partners of the polycystins both in cilia and at adhesion complexes and demonstrate RPTPγ phosphatase activity is central to the molecular mechanisms governing polycystin-dependent signaling. This article is part of a Special Issue entitled: Polycystic Kidney Disease.
Collapse
Affiliation(s)
- Catherine A Boucher
- Department of Medical Genetics, Cambridge Institute for Medical Research, Cambridge, CB2 2XY, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Pietrzak-Nowacka M, Safranow K, Byra E, Nowosiad M, Marchelek-Myśliwiec M, Ciechanowski K. Glucose metabolism parameters during an oral glucose tolerance test in patients with autosomal dominant polycystic kidney disease. Scandinavian Journal of Clinical and Laboratory Investigation 2010; 70:561-7. [DOI: 10.3109/00365513.2010.527012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
33
|
Lee YJ, Chen HY, Hsu WL, Ou CM, Wong ML. Diagnosis of feline polycystic kidney disease by a combination of ultrasonographic examination and PKD1
gene analysis. Vet Rec 2010; 167:614-8. [DOI: 10.1136/vr.c4605] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Y-J. Lee
- Veterinary Medical Teaching Hospital
| | | | - W-L. Hsu
- Graduate Institute of Microbiology and Public Health; College of Veterinary Medicine; National Chung Hsing University; 250 Kuo Kuang Road Taichung 402 Taiwan
| | - C-M. Ou
- Department of Veterinary Medicine
| | | |
Collapse
|
34
|
Lee YJ, Chen HY, Wong ML, Hsu WL. Molecular detection of autosomal-dominant feline polycystic kidney disease by multiplex amplification refractory mutation system polymerase chain reaction. J Vet Diagn Invest 2010; 22:424-8. [PMID: 20453219 DOI: 10.1177/104063871002200314] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Feline autosomal-dominant polycystic kidney disease (ADPKD), with its characteristic growth of fluid-filled cysts of different sizes, is the most prevalent inherited genetic disease of cats. The point mutation (C-->A transversion) in exon 29 of the PKD1 gene is known to contribute to ADPKD development and can thus serve as a target for the molecular genetic diagnosis of ADPKD. To this end, a simple amplification refractory mutation system (ARMS) polymerase chain reaction (PCR) was designed with 3 primers: 2 forward primers specifically targeting either the mutant or normal allele, and 1 universal reverse primer for amplification of both alleles. The new method was tested on the DNA from 35 feline blood samples, which included 15 mutant cats and 20 wild type cats. As verified by direct DNA sequencing, both sensitivity and specificity of this tri-primer ARMS PCR were 100%. As the multiplex ARMS PCR test can be performed in a single PCR reaction without other post-PCR procedures, it is a simple and accurate method for molecular studies of feline ADPKD.
Collapse
Affiliation(s)
- Ya-Jane Lee
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung-Hsing University, Taichung 402, Taiwan
| | | | | | | |
Collapse
|
35
|
Bosl WJ, Li R. The role of noise and positive feedback in the onset of autosomal dominant diseases. BMC SYSTEMS BIOLOGY 2010; 4:93. [PMID: 20587063 PMCID: PMC2902440 DOI: 10.1186/1752-0509-4-93] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Accepted: 06/29/2010] [Indexed: 01/26/2023]
Abstract
Background Autosomal dominant (AD) diseases result when a single mutant or non-functioning gene is present on an autosomal chromosome. These diseases often do not emerge at birth. There are presently two prevailing theories explaining the expression of AD diseases. One explanation originates from the Knudson two-hit theory of hereditary cancers, where loss of heterozygosity or occurrence of somatic mutations impairs the function of the wild-type copy. While these somatic second hits may be sufficient for stable disease states, it is often difficult to determine if their occurrence necessarily marks the initiation of disease progression. A more direct consequence of a heterozygous genetic background is haploinsufficiency, referring to a lack of sufficient gene function due to reduced wild-type gene copy number; however, haploinsufficiency can involve a variety of additional mechanisms, such as noise in gene expression or protein levels, injury and second hit mutations in other genes. In this study, we explore the possible contribution to the onset of autosomal dominant diseases from intrinsic factors, such as those determined by the structure of the molecular networks governing normal cellular physiology. Results First, simple models of single gene insufficiency using the positive feedback loops that may be derived from a three-component network were studied by computer simulation using Bionet software. The network structure is shown to affect the dynamics considerably; some networks are relatively stable even when large stochastic variations in are present, while others exhibit switch-like dynamics. In the latter cases, once the network switches over to the disease state it remains in that state permanently. Model pathways for two autosomal dominant diseases, AD polycystic kidney disease and mature onset diabetes of youth (MODY) were simulated and the results are compared to known disease characteristics. Conclusions By identifying the intrinsic mechanisms involved in the onset of AD diseases, it may be possible to better assess risk factors as well as lead to potential new drug targets. To illustrate the applicability of this study of pathway dynamics, we simulated the primary pathways involved in two autosomal dominant diseases, Polycystic Kidney Disease (PKD) and mature onset diabetes of youth (MODY). Simulations demonstrate that some of the primary disease characteristics are consistent with the positive feedback - stochastic variation theory presented here. This has implications for new drug targets to control these diseases by blocking the positive feedback loop in the relevant pathways.
Collapse
|
36
|
Bartolini F, Gundersen GG. Formins and microtubules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1803:164-73. [PMID: 19631698 DOI: 10.1016/j.bbamcr.2009.07.006] [Citation(s) in RCA: 135] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2009] [Revised: 05/29/2009] [Accepted: 07/04/2009] [Indexed: 02/06/2023]
Abstract
Formins have recently been recognized as prominent regulators of the microtubule (MT) cytoskeleton where they modulate the dynamics of selected MTs in interphase and mitosis. The association of formins with the MT cytoskeleton and their action on MT dynamics are relatively unexplored areas, yet growing evidence supports a direct role in their regulation of MT stability independent of their activity on actin. Formins regulate MT stability alone or in combination with accessory MT binding proteins that have previously been implicated in the stabilization of MTs downstream of polarity cues. As actin and MT arrays are typically remodeled downstream of signaling pathways that orchestrate cell shape and division, formins are emerging as excellent candidates for coordinating the responses of the cytoskeletal in diverse regulated and homeostatic processes.
Collapse
Affiliation(s)
- F Bartolini
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | | |
Collapse
|
37
|
McKnight AJ, O'Donoghue D, Peter Maxwell A. Annotated chromosome maps for renal disease. Hum Mutat 2009; 30:314-20. [PMID: 19085929 DOI: 10.1002/humu.20885] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
A combination of linkage analyses and association studies are currently employed to promote the identification of genetic factors contributing to inherited renal disease. We have standardized and merged complex genetic data from disparate sources, creating unique chromosomal maps to enhance genetic epidemiological investigations. This database and novel renal maps effectively summarize genomic regions of suggested linkage, association, or chromosomal abnormalities implicated in renal disease. Chromosomal regions associated with potential intermediate clinical phenotypes have been integrated, adding support for particular genomic intervals. More than 500 reports from medical databases, published scientific literature, and the World Wide Web were interrogated for relevant renal-related information. Chromosomal regions highlighted for prioritized investigation of renal complications include 3q13-26, 6q22-27, 10p11-15, 16p11-13, and 18q22. Combined genetic and physical maps are effective tools to organize genetic data for complex diseases. These renal chromosome maps provide insights into renal phenotype-genotype relationships and act as a template for future genetic investigations into complex renal diseases. New data from individual researchers and/or future publications can be readily incorporated to this resource via a user-friendly web-form accessed from the website: www.qub.ac.uk/neph-res/CORGI/index.php.
Collapse
Affiliation(s)
- Amy Jayne McKnight
- Nephrology Research Group, Queen's University of Belfast, United Kingdom.
| | | | | |
Collapse
|
38
|
Pretransplant genetic testing of live kidney donors at risk for autosomal dominant polycystic kidney disease. Transplantation 2009; 87:6-7. [PMID: 19136884 DOI: 10.1097/tp.0b013e318191965d] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Abstract
Polycystic kidney diseases (PKDs) represent a large group of progressive renal disorders characterized by the development of renal cysts leading to end-stage renal disease. Enormous strides have been made in understanding the pathogenesis of PKDs and the development of new therapies. Studies of autosomal dominant and recessive polycystic kidney diseases converge on molecular mechanisms of cystogenesis, including ciliary abnormalities and intracellular calcium dysregulation, ultimately leading to increased proliferation, apoptosis and dedifferentiation. Here we review the pathobiology of PKD, highlighting recent progress in elucidating common molecular pathways of cystogenesis. We discuss available models and challenges for therapeutic discovery as well as summarize the results from preclinical experimental treatments targeting key disease-specific pathways.
Collapse
|
40
|
Masoumi A, Reed-Gitomer B, Kelleher C, Bekheirnia MR, Schrier RW. Developments in the management of autosomal dominant polycystic kidney disease. Ther Clin Risk Manag 2008; 4:393-407. [PMID: 18728845 PMCID: PMC2504069 DOI: 10.2147/tcrm.s1617] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent life- threatening, hereditary disease. ADPKD is more common than sickle cell anemia, cystic fibrosis, muscular dystrophy, hemophilia, Down's syndrome, and Huntington's disease combined. ADPKD is a multisystemic disorder characterized by the progressive development of renal cysts and marked renal enlargement. Structural and functional renal deterioration occurs in ADPKD patients and is the fourth leading cause of end-stage renal disease (ESRD) in adults. Aside from the renal manifestations, extrarenal structural abnormalities, such as liver cysts, cardiovascular abnormalities, and intracranial aneurysms may lead to morbidity and mortality. Recent studies have identified prognostic factors for progressive renal impairment including gender, race, age, proteinuria, hematuria, hypertension and increased left ventricular mass index (LVMI). Early diagnosis and better understanding of the pathophysiology of the disease provides the opportunity to aggressivly treat hypertension with renin-angiotensin-aldosterone system inhibitors and thereby potentially reduce LVMI, prevent cardiovascular morbidity and mortality and slow progression of the renal disease.
Collapse
Affiliation(s)
- Amirali Masoumi
- Department of Medicine, Health Sciences Center, University of Colorado School of Medicine Denver, CO 80262, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Primary (nonmotile) cilia are currently enjoying a renaissance in light of novel ascribed functions ranging from mechanosensory to signal transduction. Their importance for key developmental pathways such as Sonic Hedgehog (Shh) and Wnt is beginning to emerge. The function of nodal cilia, for example, is vital for breaking early embryonic symmetry, Shh signaling is important for tissue morphogenesis and successful Wnt signaling for organ growth and differentiation. When ciliary function is perturbed, photoreceptors may die, kidney tubules develop cysts, limb digits multiply and brains form improperly. The etiology of several uncommon disorders has recently been associated with cilia dysfunction. The causative genes are often similar and their cognate proteins certainly share cellular locations and/or pathways. Animal models of ciliary gene ablation such as Ift88, Kif3a, and Bbs have been invaluable for understanding the broad function of the cilium. Herein, we describe the wealth of information derived from the study of the ciliopathies and their animal models.
Collapse
|
42
|
Autosomal dominant polycystic kidney disease is not a risk factor for post-transplant diabetes mellitus. Matched-pair design multicenter study. Arch Med Res 2007; 39:312-9. [PMID: 18279704 DOI: 10.1016/j.arcmed.2007.10.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2007] [Accepted: 10/15/2007] [Indexed: 01/14/2023]
Abstract
BACKGROUND Post-transplant diabetes mellitus (PTDM) is a relatively common complication of kidney transplantation. The aim of our work was to compare the incidence of PTDM in kidney transplant recipients with and without autosomal dominant polycystic kidney disease (ADPKD) in a matched-pair design study. METHODS In total, 98 pairs of graft recipients, all of Caucasian origin and who received a kidney from the same cadaveric donor, were included in the study. The following clinical data were collected for statistical analysis: age, body mass index (BMI) before transplant, length and type of dialysis treatment, residual diuresis, and cold and warm graft ischemia time. Diabetes was diagnosed based on American Diabetes Association (ADA) criteria. RESULTS Incidence of PTDM was 19.4% in the ADPKD group and 18.4% in the non-ADPKD group, with no significant differences between groups. Multivariate logistic regression analysis of the PTDM risk in the ADPKD group including age, gender, BMI, and dialysis time as independent variables indicated that only higher residual diuresis is a significant independent risk factor (OR = 5.64 per every L/24 h, 95% CI = 1.31-24.33, p = 0.017). Similarly, logistic regression analysis adjusted for age and gender in the non-ADPKD group has shown that significant independent risk factors are BMI (OR = 1.30 per every kg/m(2), 95% CI = 1.06-1.59, p = 0.0094), longer dialysis time prior to transplant (OR = 1.036 per each month, 95% CI = 1.004-1.070, p = 0.025), and a history of arterial hypertension (OR = 9.09, 95% CI = 1.20-68.66, p = 0.030). CONCLUSIONS In this paired analysis, our results suggest that diagnosis of ADPKD does not increase risk of PTDM.
Collapse
|
43
|
Boyer O, Gagnadoux MF, Guest G, Biebuyck N, Charbit M, Salomon R, Niaudet P. Prognosis of autosomal dominant polycystic kidney disease diagnosed in utero or at birth. Pediatr Nephrol 2007; 22:380-8. [PMID: 17124604 DOI: 10.1007/s00467-006-0327-8] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2006] [Revised: 09/12/2006] [Accepted: 09/12/2006] [Indexed: 11/24/2022]
Abstract
The use of prenatal ultrasonography has resulted in increased numbers of fetuses being diagnosed with autosomal dominant polycystic kidney disease (ADPKD), but the long-term prognosis is still not well-known. Between 1981 and 2006 we followed 26 consecutive children with enlarged hyperechoic kidneys detected between the 12th week of pregnancy and the first day of life (Day 1) as well as one affected parent. Three other fetuses were excluded following the termination of the pregnancy. The mother was the transmitting parent in 16 of the 26 children (ns, p=0.1). Clinical features that presented during follow-up were oligoamnios (5/26), neonatal pneumothorax (3/26), pyelonephritis (5/26), gross hematuria (2/26), hypertension (5/26), proteinuria (2/26) and chronic renal insufficiency (CRI) (2/26). At the last follow-up (mean duration of follow-up: 76 months; range: 0.5-262 months), 19 children (mean age: 5.5 years) were asymptomatic, five (mean age: 8.5 years) had hypertension, two (mean age: 9.7 years) had proteinuria and two (mean age: 19 years) had CRI. Children presenting enlarged kidneys postnatally tended to have more clinical manifestations than their counterparts who did not. Of 25 siblings of the patients, seven had renal cysts; these were detected during childhood in five siblings and in utero in two siblings. In conclusion, prognosis is favourable in most children with prenatal ADPKD, at least during childhood. The sex of the transmitting parent is not a risk factor of prenatal ADPKD. A high proportion of siblings develop early renal cysts. Abnormalities visualized by ultrasonography appear to be associated to more clinical manifestations.
Collapse
Affiliation(s)
- Olivia Boyer
- Service de Néphrologie Pédiatrique, Hôpital Necker Enfants Malades, 149 rue de Sèvres, 75743, Paris cedex 15, France
| | | | | | | | | | | | | |
Collapse
|
44
|
Yoo KH, Sung YH, Yang MH, Jeon JO, Yook YJ, Woo YM, Lee HW, Park JH. Inactivation of Mxi1 induces Il-8 secretion activation in polycystic kidney. Biochem Biophys Res Commun 2007; 356:85-90. [PMID: 17350592 DOI: 10.1016/j.bbrc.2007.02.103] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2007] [Accepted: 02/16/2007] [Indexed: 10/23/2022]
Abstract
The Mxi1 proteins are biochemical and biological antagonists of c-myc oncoprotein. It has been reported that the overexpression pattern of c-myc might be similar to a molecular feature of early and late stages of human autosomal dominant polycystic kidney disease. We identified the cyst phenotype in Mxi1-deficient mice aged 6-12 months using H&E staining. Some chemokines containing a protein domain similar to human IL-8, which is associated with the inflammatory response, were subsequently selected from the up-regulated genes. We confirmed the expression level of these chemokines and measured protein concentrations of IL-8 using ELISA in the Mxi1-knockdown cells. IL-8 was found to be significantly increased in Mxi1-knockdown cells. We found that p38 MAP kinase activation was involved in the signal transduction of the Mxi1-inactivated secretion of IL-8. Therefore, we could suggest that the inactivation of Mxi1 leads to the inflammatory response and has the potential to induce polycystic renal disease.
Collapse
Affiliation(s)
- Kyung Hyun Yoo
- Department of Biological Science, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Freichel M, Flockerzi V. Biological functions of TRPs unravelled by spontaneous mutations and transgenic animals. Biochem Soc Trans 2007; 35:120-3. [PMID: 17233616 DOI: 10.1042/bst0350120] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The identification of the biological functions of TRP (transient receptor potential) proteins requires genetic approaches because a selective TRP channel pharmacology to unravel the roles of TRPs is not available so far for most TRPs. A survey is therefore presented of transgenic animal models carrying mutations in TRP genes, as well as of those TRP genes that when mutated result in human disease; the chromosomal locations of TRP channel genes in the human and mouse are also presented.
Collapse
Affiliation(s)
- M Freichel
- Experimentelle und Klinische Pharmakologie und Toxikologie, Gebäude 46, Medizinische Fakultät, Universität des Saarlandes, 66421 Homburg, Germany.
| | | |
Collapse
|
46
|
Binczak-Kuleta A, Rozanski J, Domanski L, Myslak M, Ciechanowski K, Ciechanowicz A. DNA microsatellite analysis in families with autosomal dominant polycystic kidney disease (ADPKD): the first Polish study. J Appl Genet 2006; 47:383-9. [PMID: 17132904 DOI: 10.1007/bf03194649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common inherited renal disorders with genetic heterogeneity. Mutations of two known genes are responsible for this disease: PKD1 at 16p13.3 and PKD2 at 4q21-23. A majority of cases (85%) are caused by mutations in PKD1. Because direct mutation screening remains complex, we describe here the application of an efficient approach to studies based on highly informative dinucleotide and tetranucleotide repeats flanking genes PKD1 and PKD2. METHODS For this study a series of microsatellites closely linked to locus PKD1 (D16S291, D16S663, D16S665, D16S283, D16S407, D16S475) and to locus PKD2 (D4S1563, D4S2929, D4S414, D4S1534, D4S423) were selected. Short (81-242 bp) DNA fragments containing the tandem repeats were amplified by polymerase chain reaction (PCR). The number of repeat units of microsatelite markers was determined by fluorescent capillary electrophoresis. RESULTS DNA microsatellite analysis was performed in 25 Polish ADPKD families and established the type of disease (21 families PKD1-type, 1 family PKD2-type). CONCLUSIONS While a disease-causing mutation in the PKD1 and PKD2 genes cannot be identified, DNA microsatellite analysis provided an early diagnosis and may be considered in ADPKD families.
Collapse
Affiliation(s)
- Agnieszka Binczak-Kuleta
- Pomeranian Medical University, Department of Laboratory Diagnostics & Molecular Medicine, Szczecin, Poland.
| | | | | | | | | | | |
Collapse
|
47
|
Pietrzak-Nowacka M, Rózanski J, Safranow K, Kedzierska K, Dutkiewicz G, Ciechanowski K. Autosomal Dominant Polycystic Kidney Disease Reduces the Risk of Diabetes Mellitus. Arch Med Res 2006; 37:360-4. [PMID: 16513485 DOI: 10.1016/j.arcmed.2005.07.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2005] [Accepted: 07/11/2005] [Indexed: 11/22/2022]
Abstract
BACKGROUND Patients with autosomal dominant polycystic kidney disease (ADPKD) are at greater risk of new-onset diabetes after transplantation as compared with other renal graft recipients. METHODS We mailed questionnaires to 459 ADPKD patients retrieved from the Polish Registry of ADPKD. We analyzed data from 291 respondents and 271 siblings with a known status of ADPKD and diabetes. RESULTS The prevalence of transplant-unrelated diabetes was significantly higher in siblings without ADPKD (8.2%) than in respondents (1.7%; p = 0.0028) and their ADPKD siblings (2.0%; p = 0.023). Univariate logistic regression demonstrated that the prevalence odds ratio (POR) for transplant-unrelated diabetes in the pooled ADPKD group vs. siblings without ADPKD was 0.21 (95% CI: 0.08-0.54, p = 0.0013). Multivariate regression accounting for age and gender disclosed an even smaller POR for diabetes (0.18) in ADPKD patients (95% CI: 0.07-0.47, p = 0.00049). Age was a significant risk factor for diabetes (POR 1.05, 95% CI: 1.01-1.09 per year of life; p = 0.025) and gender was without effect. The prevalence of diabetes in females and males with vs. without ADPKD was similar (1.6% vs. 8.3%, p = 0.0091 for females; 2.2% vs. 8.0%, p = 0.069 for males). Age and gender were not inter-related. In the group of siblings without ADPKD diabetes was associated with higher age (62.2 +/- 15.6 vs. 47.0 +/- 16.3 years, p = 0.0053). CONCLUSIONS Our findings demonstrate lower prevalence of transplant-unrelated diabetes among ADPKD patients. We hypothesize that metabolic disturbances in polycystic kidneys suppress the synthesis of endogenous glucose and reduce renal breakdown of insulin.
Collapse
Affiliation(s)
- Maria Pietrzak-Nowacka
- Department of Nephrology, Transplantology and Internal Medicine, Pomeranian Medical University, Szczecin, Poland
| | | | | | | | | | | |
Collapse
|
48
|
Young AE, Biller DS, Herrgesell EJ, Roberts HR, Lyons LA. Feline polycystic kidney disease is linked to the PKD1 region. Mamm Genome 2005; 16:59-65. [PMID: 15674734 DOI: 10.1007/s00335-004-2412-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 08/24/2004] [Indexed: 10/25/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a commonly inherited disorder (1/1000) in humans characterized by fluid-filled cysts in the kidneys. Defects in the PKD genes, PKD1 and PKD2, cause 85% and 15% of human ADPKD cases, respectively. Mutations in the PKHD1 gene cause autosomal recessive PKD (ARPKD). Mutations in several genes, including Nek8, cause PKD in mice. Although PKD affects 38% of Persian cats worldwide, making it the most prominent inherited feline disease, a causative gene has not been identified. Feline PKD is an autosomal dominant disease with clinical presentations similar to human ADPKD. Forty-three microsatellites were chosen from the feline genetic maps based on known homology with human chromosomal regions containing the PKD1, PKD2, PKHD1, and Nek8 genes. Linkage analysis using seven Persian cat pedigrees segregating for PKD has shown significant linkage and no recombinants (Z=5.83, theta=0) between the PKD disease phenotype and marker FCA476, which is within 10 cR of the feline PKD1 gene on Chromosome E3. This suggests that the PKD1 gene or another gene within this region may cause feline PKD. Further investigation into the cause of PKD will be valuable for feline health and provide insights into human ADPKD.
Collapse
Affiliation(s)
- Amy E Young
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California, 1114 Tupper Hall, Davis, California, 95616, USA,
| | | | | | | | | |
Collapse
|
49
|
Peltola P, Lumiaho A, Miettinen R, Pihlajamäki J, Sandford R, Laakso M. Genetics and phenotypic characteristics of autosomal dominant polycystic kidney disease in Finns. J Mol Med (Berl) 2005; 83:638-46. [PMID: 15772804 DOI: 10.1007/s00109-005-0644-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2004] [Accepted: 12/03/2004] [Indexed: 11/29/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease, leading to renal insufficiency and renal transplantation. Mutation screening in the major gene for ADPKD, the polycystic kidney disease type 1 (PKD1) gene, has often been incomplete because of multiple homologous copies of this gene elsewhere on chromosome 16. Furthermore, there are only a few studies investigating genotype-phenotype correlations in patients with ADPKD. In this study, we screened the entire coding region of the PKD1 and PKD2 genes in 17 Finnish families with ADPKD via long-range polymerase chain reaction, single-strand conformation polymorphism analysis, and direct sequencing. We were able to identify mutations co-segregating with ADPKD in all 16 families linked to PKD1 by haplotype analysis. Of these mutations, six were insertions/deletions, five nonsense mutations, and five missense mutations. In the only PKD2-linked family, we found a missense mutation, R322Q. With the exception of one mutation (L845S in PKD1), all mutations were novel. Mutations and their location did not have a strong correlation with the phenotype with the exception of subarachnoidal hemorrhage or brain aneurysm, where mutations were located more often at the 5' end of the PKD1 gene than at the 3' end of the PKD1 gene.
Collapse
Affiliation(s)
- Paula Peltola
- Department of Medicine, University of Kuopio, P.O. Box 1777, 70211 Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
50
|
Velinov M, Kupferman J, Gu H, Macera MJ, Babu A, Jenkins EC, Kupchik G. Polycystic kidneys and del (4)(q21.1q21.3): further delineation of a distinct phenotype. Eur J Med Genet 2005; 48:51-5. [PMID: 15953406 DOI: 10.1016/j.ejmg.2005.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Accepted: 07/29/2004] [Indexed: 12/31/2022]
Abstract
A three year-old boy was evaluated because of growth and developmental delay, hypotonia and dysmorphic features. G-banding analysis revealed a small interstitial deletion of the long arm of chromosome four described as 46,XY,del (4)(q21.1q21.3). This patient's findings on physical exam included relative macrocephaly, frontal bossing, short fingers with clinodactyly and were consistent with the phenotypes of previously reported deletions involving the 4q21--> 4q22 band region (Am. J. Med. Genet. 68 (1997) 400-405). To date there are 10 reported live-born cases with such deletions and similar features. The case reported here delimits a minimal critical region for this phenotype to chromosomal region 4q21. Our patient was also found to have cysts in both his kidneys. The gene for type II polycystic kidney disease (PKD2) has been mapped to chromosomal region 4q21--> 4q23. FISH analysis, with a probe including the PKD2 gene, demonstrated hemizygosity at this locus. Thus the absence of one of the PKD2 alleles in the case reported here is associated with early bilateral cyst development. Kidney ultrasound/autopsy studies were reported in seven of the patients with the characteristic phenotype, and were positive for cysts in four cases including the one presented here (Clin. Genet. 31 (1987) 199-205; Am. J. Med. Genet. 68 (1997) 400-405; Am. J. Med. Genet. 40 (1991) 77-790. Our report supports the presence of a distinct phenotype associated with a deleted chromosomal region within 4q21. Hemizygosity for the PKD2 gene is likely in such deletions and may lead to renal cyst formation.
Collapse
Affiliation(s)
- M Velinov
- Department of Cytogenetics, NYS Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, New York, 10314 NY, USA.
| | | | | | | | | | | | | |
Collapse
|