201
|
Zhou X, Fan LX, Peters DJM, Trudel M, Bradner JE, Li X. Therapeutic targeting of BET bromodomain protein, Brd4, delays cyst growth in ADPKD. Hum Mol Genet 2015; 24:3982-93. [PMID: 25877301 DOI: 10.1093/hmg/ddv136] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 04/13/2015] [Indexed: 01/13/2023] Open
Abstract
In this study, we identified a BET bromodomain (BRD) protein, Brd4, not only as a novel epigenetic regulator of autosomal dominant polycystic kidney disease (ADPKD) but also as a novel client protein of Hsp90. We found that Brd4 was upregulated in Pkd1 mutant mouse renal epithelial cells and tissues. This upregulation of Brd4 appears to result from the chaperone activity of Hsp90 and escape proteasomal degradation. We further identify that Brd4 is an upstream regulator of the expression of c-Myc which has been upregulated in all rodent models of PKD and ADPKD patients with unknown mechanism. Inhibition of Brd4 in Pkd1 mutant renal epithelial cells with JQ1, a selective small-molecular inhibitor of BET BRD protein(s), (1) decreased the levels of c-Myc mRNA and protein; (2) increased the levels of p21 mRNA and protein, which was transcriptionally repressed by c-Myc; (3) decreased the phosphorylation of Rb; and (4) decreased cystic epithelial cell proliferation as shown by inhibition of S-phase entry. Most importantly, treatment with JQ1 strikingly delayed cyst growth and kidney enlargement, and preserved renal function in two early stage genetic mouse strains with Pkd1 mutations. This study not only provides one of the mechanisms of how c-Myc is upregulated in PKD but also suggests that targeting Brd4 with JQ1 may function as a novel epigenetic approach in ADPKD. The unraveled link between Brd4 and Hsp90 in ADPKD may also be a general mechanism for the upregulation of Brd4 in cancer cells and opens up avenues for combination therapies against ADPKD and cancer.
Collapse
Affiliation(s)
- Xia Zhou
- Department of Internal Medicine, Kidney Institute
| | - Lucy X Fan
- Department of Internal Medicine, Kidney Institute
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Trudel
- Molecular Genetics and Development, Institut de Recherches Cliniques de Montreal, Université de Montreal, Faculté de Médecine, Montreal, Quebec, Canada and
| | - James E Bradner
- Department of Medicine, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Xiaogang Li
- Department of Internal Medicine, Kidney Institute, Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS 66160, USA,
| |
Collapse
|
202
|
Jin S, Cui K, Sun ZQ, Shen YY, Li P, Wang ZD, Li FF, Gong KN, Li S. Screening analysis of candidate gene mutations in a kindred with polycystic liver disease. World J Gastroenterol 2015; 21:2343-2351. [PMID: 25741140 PMCID: PMC4342909 DOI: 10.3748/wjg.v21.i8.2343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 09/03/2014] [Accepted: 11/19/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To find potential mutable sites by detecting mutations of the candidate gene in a kindred with polycystic liver disease (PCLD).
METHODS: First, we chose a kindred with PCLD and obtained five venous blood samples of this kindred after the family members signed the informed consent form. In the kindred two cases were diagnosed with PCLD, and the left three cases were normal individuals. All the blood samples were preserved at -85 °C. Second, we extracted the genomic DNA from the venous blood samples of the kindred using a QIAamp DNA Mini Kit and then performed long-range polymerase chain reaction (PCR) with different primers. The exons of PKD1 were all sequenced with the forward and reverse primers to ensure the accuracy of the results. Next, we purified the PCR products and directly sequenced them using Big Dye Terminator Chemistry version 3.1. The sequencing reaction was conducted with BiomekFX (Beckman). Finally, we analyzed the results.
RESULTS: A total of 42 normal exons were identified in detecting mutations of the PKD1 gene. A synonymous mutation occurred in exon 5. The mutation was a homozygous T in the proband and was C in the reference sequence. This mutation was located in the third codon and did not change the amino acid encoded by the codon. Missense mutations occurred in exons 11 and 35. These mutations were located in the second codon; they changed the amino acid sequence and existed in the dbSNP library. A nonsense mutation occurred in exon 15. The mutation was a heterozygous CT in the proband and was C in the reference sequence. This mutation was located in the first codon and resulted in a termination codon. This mutation had an obvious influence on the encoded protein and changed the length of the protein from 4303 to 2246 amino acids. This was a new mutation that was not present in the dbSNP library.
CONCLUSION: The nonsense mutation of exon 15 existed in the proband and in the third individual. Additionally, the proband was heterozygous for this mutation, so the mutable site was a pathogenic mutation.
Collapse
|
203
|
Christou-Savina S, Beales PL, Osborn DPS. Evaluation of zebrafish kidney function using a fluorescent clearance assay. J Vis Exp 2015:e52540. [PMID: 25742415 DOI: 10.3791/52540] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The zebrafish embryo offers a tractable model to study organogenesis and model human genetic disease. Despite its relative simplicity, the zebrafish kidney develops and functions in almost the same way as humans. A major difference in the construction of the human kidney is the presence of millions of nephrons compared to the zebrafish that has only two. However, simplifying such a complex system into basic functional units has aided our understanding of how the kidney develops and operates. In zebrafish, the midline located glomerulus is responsible for the initial blood filtration into two pronephric tubules that diverge to run bilaterally down the embryonic axis before fusing to each other at the cloaca. The pronephric tubules are heavily populated by motile cilia that facilitate the movement of filtrate along the segmented tubule, allowing the exchange of various solutes before finally exiting via the cloaca. Many genes responsible for CKD, including those related to ciliogenesis, have been studied in zebrafish. However, a major draw back has been the difficulty in evaluating zebrafish kidney function after genetic manipulation. Traditional assays to measure kidney dysfunction in humans have proved non translational to zebrafish, mainly due to their aquatic environment and small size. For example, it is not physically possible to extract blood from embryonic staged fish for analysis of urea and creatinine content, as they are too small. In addition, zebrafish do not produce enough urine for testing on a simple proteinuria 'dipstick', which is often performed during initial patient examinations. We describe a fluorescent assay that utilizes the optical transparency of the zebrafish to quantitatively monitor the clearance of a fluorescent dye, over time, from the vasculature and out through the kidney, to give a read out of renal function.
Collapse
Affiliation(s)
| | - Philip L Beales
- Genetics and Genomic Medicine, Institute of Child Health, University College London
| | - Daniel P S Osborn
- Molecular Cell Science Research Centre, St. George's University of London;
| |
Collapse
|
204
|
Kazancioglu R, Gursu M. New options in the treatment of autosomal dominant polycystic kidney disease. Ren Fail 2015; 37:535-41. [PMID: 25682970 DOI: 10.3109/0886022x.2015.1013404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Autosomal dominant polycystic disease (ADPKD) is one of the most common monogenic disorders, and globally is among the most common hereditary causes of end stage kidney disease. Until recently, the causes of this disease remained obscure. However, in the past decade there have been enormous advances in the understanding of the pathophysiology and genetics of this condition, and recent studies have suggested the possibility of specific treatment for slowing cyst growth. This review will focus on the new options for the control of ADPKD.
Collapse
Affiliation(s)
- Rumeyza Kazancioglu
- Department of Nephrology, Bezmialem Vakif University School of Medicine , Istanbul , Turkey and
| | | |
Collapse
|
205
|
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: 2.1] [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
|
206
|
Bourcier R, Redon R, Desal H. Genetic investigations on intracranial aneurysm: update and perspectives. J Neuroradiol 2015; 42:67-71. [PMID: 25676693 DOI: 10.1016/j.neurad.2015.01.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 11/26/2014] [Accepted: 01/12/2015] [Indexed: 11/30/2022]
Abstract
Detection of an intracranial aneurysm (IA) is a common finding in MRI practice. Nowadays, the incidence of unruptured IA seems to be increasing with the continuous evolution of imaging techniques. Important modifiable risk factors for SAH are well defined, but familial history of IA is the best risk marker for the presence of IA. Numerous heritable conditions are associated with IA formation but these syndromes account for less than 1% of all IAs in the population. No diagnostic test based on genetic knowledge is currently available to identify theses mutations and patients who are at higher risk for developing IAs. In the longer term, a more comprehensive understanding of independent and interdependent molecular pathways germane to IA formation and rupture may guide the physician in developing targeted therapies and optimizing prognostic risk assessment.
Collapse
Affiliation(s)
- Romain Bourcier
- Department of neuroradiology, CHU Nantes, boulevard J.-Monod, 44000 Nantes, France; 1087 Inserm unit, institut du thorax, 44000 Nantes, France.
| | - Richard Redon
- 1087 Inserm unit, institut du thorax, 44000 Nantes, France
| | - Hubert Desal
- Department of neuroradiology, CHU Nantes, boulevard J.-Monod, 44000 Nantes, France; 1087 Inserm unit, institut du thorax, 44000 Nantes, France
| |
Collapse
|
207
|
Muto S, Kawano H, Higashihara E, Narita I, Ubara Y, Matsuzaki T, Ouyang J, Torres VE, Horie S. The effect of tolvaptan on autosomal dominant polycystic kidney disease patients: a subgroup analysis of the Japanese patient subset from TEMPO 3:4 trial. Clin Exp Nephrol 2015; 19:867-77. [PMID: 25663351 DOI: 10.1007/s10157-015-1086-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 01/05/2015] [Indexed: 01/28/2023]
Abstract
BACKGROUND Japan is the first country in the world to approve tolvaptan for the treatment of autosomal dominant polycystic kidney disease (ADPKD), which was based on the results of Tolvaptan Efficacy and Safety in Management of Autosomal Dominant Polycystic Kidney Disease and Its Outcomes (TEMPO) 3:4 trial. To evaluate the safety and efficacy of tolvaptan, we performed a subgroup analysis in the participating Japanese ADPKD patient population. METHODS The primary outcome was the annual rate of percentage change in the total kidney volume (TKV). The secondary endpoint was the rate of kidney function change. RESULTS The tolvaptan and placebo groups included 118 and 59 patients, respectively. The annual rate of percentage changes in TKV were 1.3 % [95 % confidence interval (CI) 0.4-2.1] in the tolvaptan group, and 5.0 % (95 % CI 3.9-6.2) in the placebo group (P < 0.001). The annual estimated glomerular filtration rate change was -3.83 mL/min/1.73 m(2) in the tolvaptan group and -5.05 mL in the placebo group for a treatment effect of +1.22 mL/min/1.73 m(2) (95 % CI 0.41-2.02: P = 0.003). Hepatic function abnormal as a serious adverse event was observed in 3 patients (2.5 %) in the tolvaptan group. CONCLUSIONS Administration of tolvaptan in the Japanese sub-population reduced the annual rate of TKV growth and slowed the rate of kidney function decline over 36 months compared to patients on placebo, thus providing a novel and effective therapy for the treatment of ADPKD. (TEMPO 3:4 ClinicalTrials.gov number, NCT00428948).
Collapse
Affiliation(s)
- Satoru Muto
- Department of Urology, Teikyo University School of Medicine, Tokyo, Japan
| | - Haruna Kawano
- Department of Urology, Juntendo University Graduate School of Medicine, Hongo 3-1-3, Bunkyo-ku, Tokyo, 113-8431, Japan
| | - Eiji Higashihara
- Department of ADPKD Research, Kyorin University School of Medicine, Tokyo, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yoshifumi Ubara
- Nephrology Center and Okinaka Memorial Institute for Medical Research, Toranomon Hospital, Tokyo, Japan
| | | | - John Ouyang
- Otsuka Pharmaceutical Development & Commercialization, Inc., Rockville, MD, USA
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, Hongo 3-1-3, Bunkyo-ku, Tokyo, 113-8431, Japan.
| |
Collapse
|
208
|
Tao S, Kakade VR, Woodgett JR, Pandey P, Suderman ED, Rajagopal M, Rao R. Glycogen synthase kinase-3β promotes cyst expansion in polycystic kidney disease. Kidney Int 2015; 87:1164-75. [PMID: 25629553 PMCID: PMC4449797 DOI: 10.1038/ki.2014.427] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/25/2014] [Accepted: 12/05/2014] [Indexed: 12/31/2022]
Abstract
Polycystic kidney diseases (PKDs) are inherited disorders characterized by the formation of fluid filled renal cysts. Elevated cAMP levels in PKDs stimulate progressive cyst enlargement involving cell proliferation and transepithelial fluid secretion often leading to end stage renal disease. The glycogen synthase kinase-3 (GSK3) family of protein kinases consists of GSK3α and GSK3β isoforms and plays a crucial role in multiple cellular signaling pathways. We previously found that GSK3β, a regulator of cell proliferation, is also crucial for cAMP generation and vasopressin mediated urine concentration by the kidneys. However, the role of GSK3β in the pathogenesis of PKDs is not known. Here we found that GSK3β expression and activity were markedly up-regulated and associated with cyst-lining epithelia in the kidneys of mice and humans with PKD. Renal collecting duct specific gene knockout of GSK3β or pharmacological inhibition of GSK3 effectively slowed the progression of PKD in mouse models of autosomal recessive or autosomal dominant PKD. GSK3 inactivation inhibited cAMP generation and cell proliferation resulting in reduced cyst expansion, improved renal function and extended lifespan. GSK3β inhibition also reduced pERK, c-Myc and Cyclin-D1, known mitogens in proliferation of cystic epithelial cells. Thus, GSK3β plays a novel functional role in PKD pathophysiology and its inhibition may be therapeutically useful to slow cyst expansion and progression of PKD.
Collapse
Affiliation(s)
- Shixin Tao
- Department of Medicine, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Vijayakumar R Kakade
- Department of Medicine, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - James R Woodgett
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital and Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Pankaj Pandey
- Department of Medicine, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Erin D Suderman
- Department of Medicine, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Madhumitha Rajagopal
- Department of Medicine, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Reena Rao
- Department of Medicine, The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
209
|
Wang W, Li F, Sun Y, Lei L, Zhou H, Lei T, Xia Y, Verkman AS, Yang B. Aquaporin-1 retards renal cyst development in polycystic kidney disease by inhibition of Wnt signaling. FASEB J 2015; 29:1551-63. [PMID: 25573755 DOI: 10.1096/fj.14-260828] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 12/15/2014] [Indexed: 01/04/2023]
Abstract
Water channel aquaporin-1 (AQP1) is expressed at epithelial cell plasma membranes in renal proximal tubules and thin descending limb of Henle. Recently, AQP1 was reported to interact with β-catenin. Here we investigated the relationship between AQP1 and Wnt signaling in in vitro and in vivo models of autosomal dominant polycystic kidney disease (PKD). AQP1 overexpression decreased β-catenin and cyclinD1 expression, suggesting down-regulation of Wnt signaling, and coimmunoprecipitation showed AQP1 interaction with β-catenin, glycogen synthase kinase 3β, LRP6, and Axin1. AQP1 inhibited cyst development and promoted branching in matrix-grown MDCK cells. In embryonic kidney cultures, AQP1 deletion increased cyst development by up to ∼ 40%. Kidney size and cyst number were significantly greater in AQP1-null PKD mice than in AQP1-expressing PKD mice, with the difference mainly attributed to a greater number of proximal tubule cysts. Biochemical analysis revealed decreased β-catenin phosphorylation and increased β-catenin expression in AQP1-null PKD mice, suggesting enhanced Wnt signaling. These results implicate AQP1 as a novel determinant in renal cyst development that may involve inhibition of Wnt signaling by an AQP1-macromolecular signaling complex.
Collapse
Affiliation(s)
- Weiling Wang
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Fei Li
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Yi Sun
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Lei Lei
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Hong Zhou
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Tianluo Lei
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Yin Xia
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - A S Verkman
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| | - Baoxue Yang
- *Department of Pharmacology, School of Basic Medical Sciences, Peking University, and State Key Laboratory of Natural and Biomimetic Drugs, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China; and Departments of Medicine and Physiology, University of California, San Francisco, San Francisco, California USA
| |
Collapse
|
210
|
ARPKD and early manifestations of ADPKD: the original polycystic kidney disease and phenocopies. Pediatr Nephrol 2015; 30:15-30. [PMID: 24584572 PMCID: PMC4240914 DOI: 10.1007/s00467-013-2706-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 11/11/2013] [Accepted: 11/12/2013] [Indexed: 12/11/2022]
Abstract
Renal cysts are clinically and genetically heterogeneous conditions. Polycystic kidney disease (PKD) is common and its characterization has paved the way for the identification of a growing number of cilia-related disorders (ciliopathies) of which most show cystic kidneys. While the recessive form of PKD (ARPKD) virtually always presents in childhood, early onset can, in some instances, also occur in the dominant form (ADPKD). Both ADPKD genes (PKD1 and PKD2) can also be inherited in a recessive way, making the story more complex with evidence for a dosage-sensitive network. Several phenocopies are known, and mutations in HNF1ß or genes that typically cause other ciliopathies, such as nephronophthisis, Bardet-Biedl, Joubert syndrome and related disorders, can mimic PKD. An accurate genetic diagnosis is crucial for genetic counseling, prenatal diagnostics, and the clinical management of patients and their families. The increasing number of genes that have to be considered in patients with cystic kidney disease is challenging to address by conventional techniques and largely benefits from next-generation sequencing-based approaches. The parallel analysis of targeted genes considerably increases the detection rate, allows for better interpretation of identified variants, and avoids genetic misdiagnoses.
Collapse
|
211
|
Schrier RW, Abebe KZ, Perrone RD, Torres VE, Braun WE, Steinman TI, Winklhofer FT, Brosnahan G, Czarnecki PG, Hogan MC, Miskulin DC, Rahbari-Oskoui FF, Grantham JJ, Harris PC, Flessner MF, Bae KT, Moore CG, Chapman AB. Blood pressure in early autosomal dominant polycystic kidney disease. N Engl J Med 2014; 371:2255-66. [PMID: 25399733 PMCID: PMC4343258 DOI: 10.1056/nejmoa1402685] [Citation(s) in RCA: 325] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Hypertension is common in autosomal dominant polycystic kidney disease (ADPKD) and is associated with increased total kidney volume, activation of the renin-angiotensin-aldosterone system, and progression of kidney disease. METHODS In this double-blind, placebo-controlled trial, we randomly assigned 558 hypertensive participants with ADPKD (15 to 49 years of age, with an estimated glomerular filtration rate [GFR] >60 ml per minute per 1.73 m(2) of body-surface area) to either a standard blood-pressure target (120/70 to 130/80 mm Hg) or a low blood-pressure target (95/60 to 110/75 mm Hg) and to either an angiotensin-converting-enzyme inhibitor (lisinopril) plus an angiotensin-receptor blocker (telmisartan) or lisinopril plus placebo. The primary outcome was the annual percentage change in the total kidney volume. RESULTS The annual percentage increase in total kidney volume was significantly lower in the low-blood-pressure group than in the standard-blood-pressure group (5.6% vs. 6.6%, P=0.006), without significant differences between the lisinopril-telmisartan group and the lisinopril-placebo group. The rate of change in estimated GFR was similar in the two medication groups, with a negative slope difference in the short term in the low-blood-pressure group as compared with the standard-blood-pressure group (P<0.001) and a marginally positive slope difference in the long term (P=0.05). The left-ventricular-mass index decreased more in the low-blood-pressure group than in the standard-blood-pressure group (-1.17 vs. -0.57 g per square meter per year, P<0.001); urinary albumin excretion was reduced by 3.77% with the low-pressure target and increased by 2.43% with the standard target (P<0.001). Dizziness and light-headedness were more common in the low-blood-pressure group than in the standard-blood-pressure group (80.7% vs. 69.4%, P=0.002). CONCLUSIONS In early ADPKD, the combination of lisinopril and telmisartan did not significantly alter the rate of increase in total kidney volume. As compared with standard blood-pressure control, rigorous blood-pressure control was associated with a slower increase in total kidney volume, no overall change in the estimated GFR, a greater decline in the left-ventricular-mass index, and greater reduction in urinary albumin excretion. (Funded by the National Institute of Diabetes and Digestive and Kidney Diseases and others; HALT-PKD [Study A] ClinicalTrials.gov number, NCT00283686.).
Collapse
Affiliation(s)
- Robert W Schrier
- From the University of Colorado, Denver (R.W.S., G.B.); University of Pittsburgh School of Medicine, Pittsburgh (K.Z.A., K.T.B., C.G.M.); Tufts Medical Center (R.D.P., D.C.M.) and Beth Israel Deaconess Medical Center (T.I.S., P.G.C.) - both in Boston; Mayo Clinic College of Medicine, Rochester, MN (V.E.T., M.C.H., P.C.H.); Cleveland Clinic, Cleveland (W.E.B.); Kansas University Medical Center, Kansas City (F.T.W., J.J.G.); Emory University School of Medicine, Atlanta (F.F.R.-O., A.B.C.); and the National Institutes of Health, Bethesda, MD (M.F.F.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
212
|
Xu JX, Lu TS, Li S, Wu Y, Ding L, Denker BM, Bonventre JV, Kong T. Polycystin-1 and Gα12 regulate the cleavage of E-cadherin in kidney epithelial cells. Physiol Genomics 2014; 47:24-32. [PMID: 25492927 DOI: 10.1152/physiolgenomics.00090.2014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Interaction of polycystin-1 (PC1) and Gα12 is important for development of kidney cysts in autosomal dominant polycystic kidney disease (ADPKD). The integrity of cell polarity and cell-cell adhesions (mainly E-cadherin-mediated adherens junction) is altered in the renal epithelial cells of ADPKD. However, the key signaling pathway for this alteration is not fully understood. Madin-Darby canine kidney (MDCK) cells maintain the normal integrity of epithelial cell polarity and adherens junctions. Here, we found that deletion of Pkd1 increased activation of Gα12, which then promoted the cystogenesis of MDCK cells. The morphology of these cells was altered after the activation of Gα12. By using liquid chromatography-mass spectrometry, we found several proteins that could be related this change in the extracellular milieu. E-cadherin was one of the most abundant peptides after active Gα12 was induced. Gα12 activation or Pkd1 deletion increased the shedding of E-cadherin, which was mediated via increased ADAM10 activity. The increased shedding of E-cadherin was blocked by knockdown of ADAM10 or specific ADAM10 inhibitor GI254023X. Pkd1 deletion or Gα12 activation also changed the distribution of E-cadherin in kidney epithelial cells and caused β-catenin to shift from cell membrane to nucleus. Finally, ADAM10 inhibitor, GI254023X, blocked the cystogenesis induced by PC1 knockdown or Gα12 activation in renal epithelial cells. Our results demonstrate that the E-cadherin/β-catenin signaling pathway is regulated by PC1 and Gα12 via ADAM10. Specific inhibition of this pathway, especially ADAM10 activity, could be a novel therapeutic regimen for ADPKD.
Collapse
Affiliation(s)
- Jen X Xu
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tzong-Shi Lu
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Suyan Li
- Division of Basic Neuroscience, McLean Hospital, Belmont, Massachusetts
| | - Yong Wu
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Lai Ding
- Harvard NeuroDiscovery Center, Boston, Massachusetts; and
| | - Bradley M Denker
- Beth Israel Deaconess Medical Center, Boston, Massachusetts; Harvard Medical School, Boston, Massachusetts
| | - Joseph V Bonventre
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Tianqing Kong
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts;
| |
Collapse
|
213
|
Smithline ZB, Nikonova AS, Hensley HH, Cai KQ, Egleston BL, Proia DA, Seeger-Nukpezah T, Golemis EA. Inhibiting heat shock protein 90 (HSP90) limits the formation of liver cysts induced by conditional deletion of Pkd1 in mice. PLoS One 2014; 9:e114403. [PMID: 25474361 PMCID: PMC4256400 DOI: 10.1371/journal.pone.0114403] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2014] [Accepted: 11/06/2014] [Indexed: 11/18/2022] Open
Abstract
Polycystic liver disease (PLD) occurs in 75-90% of patients affected by autosomal dominant polycystic kidney disease (ADPKD), which affects 1∶400-1,000 adults and arises from inherited mutations in the PKD1 or PKD2 genes. PLD can lead to bile duct obstructions, infected or bleeding cysts, and hepatomegaly, which can diminish quality of life. At present, no effective, approved therapy exists for ADPKD or PLD. We recently showed that inhibition of the molecular chaperone heat shock protein 90 (HSP90) with a small molecule inhibitor, STA-2842, induced the degradation of multiple HSP90-dependent client proteins that contribute to ADPKD pathogenesis and slowed the progression of renal cystogenesis in mice with conditional deletion of Pkd1. Here, we analyzed the effects of STA-2842 on liver size and cystic burden in Pkd-/- mice with established PLD. Using magnetic resonance imaging over time, we demonstrate that ten weeks of STA-2842 treatment significantly reduced both liver mass and cystic index suggesting selective elimination of cystic tissue. Pre-treatment cystic epithelia contain abundant HSP90; the degree of reduction in cysts was accompanied by inhibition of proliferation-associated signaling proteins EGFR and others, and induced cleavage of caspase 8 and PARP1, and correlated with degree of HSP90 inhibition and with inactivation of ERK1/2. Our results suggest that HSP90 inhibition is worth further evaluation as a therapeutic approach for patients with PLD.
Collapse
Affiliation(s)
- Zachary B. Smithline
- Yale University, New Haven, Connecticut, 06520, United States of America
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
| | - Anna S. Nikonova
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
| | - Harvey H. Hensley
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
| | - Kathy Q. Cai
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
| | - Brian L. Egleston
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
| | - David A. Proia
- Synta Pharmaceuticals, Lexington, Massachusetts, 02421, United States of America
| | - Tamina Seeger-Nukpezah
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
- Department I of Internal Medicine, Center for Integrated Oncology, University Hospital of Cologne, Cologne, 50937, Germany
| | - Erica A. Golemis
- Program in Molecular Therapeutics, Fox Chase Cancer Center, Philadelphia, Pennsylvania, 19111, United States of America
| |
Collapse
|
214
|
Intermediate conductance, Ca2+-activated K+ channels: a novel target for chronic renal diseases. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/s11515-014-1339-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
215
|
Tan M, Wettersten HI, Chu K, Huso DL, Watnick T, Friedlander S, Landesman Y, Weiss RH. Novel inhibitors of nuclear transport cause cell cycle arrest and decrease cyst growth in ADPKD associated with decreased CDK4 levels. Am J Physiol Renal Physiol 2014; 307:F1179-86. [PMID: 25234309 PMCID: PMC4254973 DOI: 10.1152/ajprenal.00406.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 09/16/2014] [Indexed: 02/07/2023] Open
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is a progressive, proliferative renal disease. Kidneys from ADPKD patients are characterized by the presence of cysts that are marked by enhanced proliferation and apoptosis of renal tubular epithelial cells. Current treatment of this disease is supportive, as there are few if any clinically validated targeted therapeutics. Given the parallels between cystic disease and cancer, and in light of our findings of the efficacy of the nuclear transport inhibitors in kidney cancer, which has similarities to ADPKD, we asked whether such inhibitors show utility in ADPKD. In this study, we tested selective inhibitors of nuclear export (SINE) in two human ADPKD cell lines and in an in vivo mouse model of ADPKD. After effective downregulation of a nuclear exporter, exportin 1 (XPO1), with KPT-330, both cell lines showed dose-dependent inhibition of cell proliferation through G₀/G₁ arrest associated with downregulation of CDK4, with minimal apoptosis. To analyze mechanisms of CDK4 decrease by XPO1 inhibition, localization of various XPO1 target proteins was examined, and C/EBPβ was found to be localized in the nucleus by XPO1 inhibition, resulting in an increase of C/EBPα, which activates degradation of CDK4. Furthermore, inhibition of XPO1 with the parallel inhibitor KPT-335 attenuated cyst growth in vivo in the PKD1 mutant mouse model Pkd1(v/v). Thus, inhibition of nuclear export by KPT-330, which has shown no adverse effects in renal serum chemistries and urinalyses in animal models, and which is already in phase 1 trials for cancers, will be rapidly translatable to human ADPKD.
Collapse
Affiliation(s)
| | | | - Kristy Chu
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David L Huso
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Terry Watnick
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | | | | | - Robert H Weiss
- Graduate Group in Comparative Pathology, Division of Nephrology, Department of Internal Medicine, and Cancer Center, University of California, Davis, California; Medical Service, Sacramento Veterans Affairs Medical Center, Sacramento, California
| |
Collapse
|
216
|
Liu W, Chen M, Wei J, He W, Li Z, Sun X, Shi Y. Modification of PCR conditions and design of exon-specific primers for the efficient molecular diagnosis of PKD1 mutations. Kidney Blood Press Res 2014; 39:536-45. [PMID: 25531466 DOI: 10.1159/000368464] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/21/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS Autosomal-dominant polycystic kidney disease (ADPKD) is a heterogeneous genetic disorder caused by mutations in the PKD1 and PKD2 genes. Currently, long-range PCR followed by nested PCR and sequencing (LRNS) is the gold standard approach for PKD1 testing. However, LRNS is complicated by the high structural and sequence complexity of PKD1, which makes the procedure for amplification and analysis of PKD1 difficult. METHODS Here in, we modified the PCR conditions and designed primers for efficient and specific amplification of both the long-range and individual exons of PKD1. RESULTS Using the modified system, seven long-range fragments were specifically amplified using two distinct sets of conditions, and all individual exon PCR assays were easily performed using a touch-down PCR method. Seven pathogenic or likely pathogenic variants, including two novel truncated frameshift indels and two novel likely pathogenic missense mutations, were identified in eight unrelated patients with or without histories of ADPKD disease (one variant was observed in two unrelated patients). Using combined bioinformatics tools, two indeterminate missense variants were identified in two sporadic patients. CONCLUSION Four novel PKD1 variants were identified in this study. We demonstrated that the modified LRNS method achieves high sensitivity and specificity for detecting pathogenic variants of ADPKD.
Collapse
Affiliation(s)
- WeiQiang Liu
- Graduate school, Southern Medical University, Guangzhou 510515, China
| | | | | | | | | | | | | |
Collapse
|
217
|
Myint TM, Rangan GK, Webster AC. Treatments to slow progression of autosomal dominant polycystic kidney disease: systematic review and meta-analysis of randomized trials. Nephrology (Carlton) 2014; 19:217-26. [PMID: 24460701 DOI: 10.1111/nep.12211] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2014] [Indexed: 01/13/2023]
Abstract
AIM Autosomal dominant polycystic kidney disease (ADPKD) is a monogenetic disorder that leads to kidney failure. Our aim was to undertake a meta-analysis of randomized trials of interventions that have been hypothesized to reduce the progression of total kidney volume (TKV) and renal function in ADPKD. METHODS Relevant trials were identified, and outcomes were: change in TKV, total cyst volume (TCV), renal function and adverse events. Meta-analysis used random effects, with results expressed as mean difference and risk ratio both with 95% confidence intervals (CI). RESULTS Eleven trials (2262 patients) were included. Compared with placebo, Target of Rapamycin complex 1 (TORC1) inhibitors (5 trials, n = 619), showed no significant change in TKV (P = 0.21), TCV (P = 0.06) or eGFR (P = 0.22). Somatostatin analogues (3 trials, n = 157) reduced TKV by 9% (95% CI -10.33 to -7.58%) but did not alter eGFR. The vasopressin receptor antagonist (n = 1455) attenuated TKV increase to 3%/year (95% CI -3.48 to -2.52) and slowed kidney function decline over a 3-year period. A single trial (n = 41) of eicosapentaenoic acid did not alter the progression of either TKV (P = 0.9) or renal dysfunction (P = 0.78). Adverse events were significant for interventions in all trials compared with placebo. CONCLUSION These data suggest that somatostatin analogues and vasopressin receptor antagonists attenuate TKV increase. The neutral effects of TORC1 inhibitors on TKV could be true, or due to heterogeneity in study population, drug efficacy and follow-up duration. In the future, further well-designed and powered trials of longer duration using new biomarkers or therapeutic agents with better tolerance are required.
Collapse
Affiliation(s)
- Thida M Myint
- Department of Renal Medicine and Transplantation, Westmead Hospital, Western Sydney Local Health District, Sydney, New South Wales, Australia
| | | | | |
Collapse
|
218
|
Petzold K, Gansevoort RT, Ong ACM, Devuyst O, Rotar L, Eckardt KU, Köttgen A, Pirson Y, Remuzzi G, Sandford R, Tesar V, Ecder T, Chaveau D, Torra R, Budde K, Le Meur Y, Wüthrich RP, Serra AL. Building a network of ADPKD reference centres across Europe: the EuroCYST initiative. Nephrol Dial Transplant 2014; 29 Suppl 4:iv26-32. [PMID: 25165183 DOI: 10.1093/ndt/gfu091] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the most common monogenic inherited kidney disease, affecting an estimated 600 000 individuals in Europe. The disease is characterized by age-dependent development of a multiple cysts in the kidneys, ultimately leading to end-stage renal failure and the need of renal replacement therapy in the majority of patients, typically by the fifth or sixth decade of life. The variable disease course, even within the same family, remains largely unexplained. Similarly, assessing disease severity and prognosis in an individual with ADPKD remains difficult. Epidemiological studies are limited due to the fragmentation of ADPKD research in Europe. METHODS The EuroCYST initiative aims: (i) to harmonize and develop common standards for ADPKD research by starting a collaborative effort to build a network of ADPKD reference centres across Europe and (ii) to establish a multicentric observational cohort of ADPKD patients. This cohort will be used to study factors influencing the rate of disease progression, disease modifiers, disease stage-specific morbidity and mortality, health economic issues and to identify predictive disease progression markers. Overall, 1100 patients will be enrolled in 14 study sites across Europe. Patients will be prospectively followed for at least 3 years. Eligible patients will not have participated in a pharmaceutical clinical trial 1 year before enrollment, have clinically proven ADPKD, an estimated glomerular filtration rate (eGFR) of 30 mL/min/1.73 m(2) and above, and be able to provide written informed consent. The baseline visit will include a physical examination and collection of blood, urine and DNA for biomarker and genetic studies. In addition, all participants will be asked to complete questionnaires detailing self-reported health status, quality of life, socioeconomic status, health-care use and reproductive planning. All subjects will undergo annual follow-up. A magnetic resonance imaging (MRI) scan will be carried out at baseline, and patients are encouraged to undergo a second MRI at 3-year follow-up for qualitative and quantitative kidney and liver assessments. CONCLUSIONS The ADPKD reference centre network across Europe and the observational cohort study will enable European ADPKD researchers to gain insights into the natural history, heterogeneity and associated complications of the disease as well as how it affects the lives of patients across Europe.
Collapse
Affiliation(s)
- Katja Petzold
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Ron T Gansevoort
- Department of Nephrology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Albert C M Ong
- Kidney Genetics Group, Academic Nephrology Unit, Department of Infection and Immunity, University of Sheffield Medical School, Sheffield, UK
| | - Olivier Devuyst
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Laura Rotar
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Kai-Uwe Eckardt
- Department of Nephrology and Hypertension, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Anna Köttgen
- Department of Internal Medicine IV, University Medical Center Freiburg, Freiburg, Germany
| | - Yves Pirson
- Department of Nephrology, Cliniques Universitaires Saint-Luc, Brussel, Belgium
| | - Giuseppe Remuzzi
- IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Bergamo, Italy and Unit of Nephrology, Dialysis and Transplantation, A.O. Papa Giovanni XXIII, Bergamo, Italy
| | - Richard Sandford
- Academic Department of Medical Genetics, University of Cambridge, Cambridge, UK
| | - Vladimir Tesar
- Department of Nephrology, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Tevfik Ecder
- Department of Nephrology, Istanbul School of Medicine, Istanbul, Turkey
| | - Dominique Chaveau
- Service de nephrologie et immunologie clinique, centre de reference des maladies renales rares (SORARE), CHU de Toulouse, universite' de Toulouse III, hopital Rangueil
| | - Roser Torra
- Inherited Kidney Diseases, Nephrology Department, Fundació Puigvert, Instituto de Investigaciones Biomédicas Sant Pau (IIB-Sant Pau), Universitat Autònoma de Barcelona, REDinREN, Instituto de Investigación Carlos III, Barcelona, Spain
| | - Klemens Budde
- Department of Nephrology, Charité Campus Mitte, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Yannick Le Meur
- Department of Nephrology, Centre Hospitalier Universitaire de Brest, Brest, France
| | - Rudolf P Wüthrich
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Andreas L Serra
- Institute of Physiology and Zurich Center for Integrative Human Physiology, University of Zurich, Zurich, Switzerland Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
219
|
Franchi F, Peterson KM, Xu R, Miller B, Psaltis PJ, Harris PC, Lerman LO, Rodriguez-Porcel M. Mesenchymal Stromal Cells Improve Renovascular Function in Polycystic Kidney Disease. Cell Transplant 2014; 24:1687-98. [PMID: 25290249 DOI: 10.3727/096368914x684619] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Polycystic kidney disease (PKD) is a common cause of end-stage renal failure, for which there is no accepted treatment. Progenitor and stem cells have been shown to restore renal function in a model of renovascular disease, a disease that shares many features with PKD. The objective of this study was to examine the potential of adult stem cells to restore renal structure and function in PKD. Bone marrow-derived mesenchymal stromal cells (MSCs, 2.5 × 10(5)) were intrarenally infused in 6-week-old PCK rats. At 10 weeks of age, PCK rats had an increase in systolic blood pressure (SBP) versus controls (126.22 ± 2.74 vs. 116.45 ± 3.53 mmHg, p < 0.05) and decreased creatinine clearance (3.76 ± 0.31 vs. 6.10 ± 0.48 µl/min/g, p < 0.01), which were improved in PKD animals that received MSCs (SBP: 114.67 ± 1.34 mmHg, and creatinine clearance: 4.82 ± 0.24 µl/min/g, p = 0.001 and p = 0.003 vs. PKD, respectively). MSCs preserved vascular density and glomeruli diameter, measured using microcomputed tomography. PCK animals had increased urine osmolality (843.9 ± 54.95 vs. 605.6 ± 45.34 mOsm, p < 0.01 vs. control), which was improved after MSC infusion and not different from control (723.75 ± 56.6 mOsm, p = 0.13 vs. control). Furthermore, MSCs reduced fibrosis and preserved the expression of proangiogenic molecules, while cyst size and number were unaltered by MSCs. Delivery of exogenous MSCs improved vascular density and renal function in PCK animals, and the benefit was observed up to 4 weeks after a single infusion. Cell-based therapy constitutes a novel approach in PKD.
Collapse
Affiliation(s)
- Federico Franchi
- Division of Cardiovascular Diseases, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | | | | | | | | | | |
Collapse
|
220
|
Wetmore JB, Calvet JP, Yu ASL, Lynch CF, Wang CJ, Kasiske BL, Engels EA. Polycystic kidney disease and cancer after renal transplantation. J Am Soc Nephrol 2014; 25:2335-41. [PMID: 24854270 PMCID: PMC4178444 DOI: 10.1681/asn.2013101122] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/15/2014] [Indexed: 01/07/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), the most common form of polycystic kidney disease (PKD), is a disorder with characteristics of neoplasia. However, it is not known whether renal transplant recipients with PKD have an increased risk of cancer. Data from the Scientific Registry of Transplant Recipients, which contains information on all solid organ transplant recipients in the United States, were linked to 15 population-based cancer registries in the United States. For PKD recipients, we compared overall cancer risk with that in the general population. We also compared cancer incidence in PKD versus non-PKD renal transplant recipients using Poisson regression, and we determined incidence rate ratios (IRRs) adjusted for age, sex, race/ethnicity, dialysis duration, and time since transplantation. The study included 10,166 kidney recipients with PKD and 107,339 without PKD. Cancer incidence in PKD recipients was 1233.6 per 100,000 person-years, 48% higher than expected in the general population (standardized incidence ratio, 1.48; 95% confidence interval [95% CI], 1.37 to 1.60), whereas cancer incidence in non-PKD recipients was 1119.1 per 100,000 person-years. The unadjusted incidence was higher in PKD than in non-PKD recipients (IRR, 1.10; 95% CI, 1.01 to 1.20). However, PKD recipients were older (median age at transplantation, 51 years versus 45 years for non-PKD recipients), and after multivariable adjustment, cancer incidence was lower in PKD recipients than in others (IRR, 0.84; 95% CI, 0.77 to 0.91). The reason for the lower cancer risk in PKD recipients is not known but may relate to biologic characteristics of ADPKD or to cancer risk behaviors associated with ADPKD.
Collapse
Affiliation(s)
- James B Wetmore
- Department of Medicine, Division of Nephrology, The Kidney Institute,
| | - James P Calvet
- The Kidney Institute, Department of Biochemistry and Molecular Biology, and Department of Cancer Biology, University of Kansas School of Medicine, Kansas City, Kansas
| | - Alan S L Yu
- Department of Medicine, Division of Nephrology, The Kidney Institute
| | - Charles F Lynch
- Department of Epidemiology, University of Iowa, Iowa City, Iowa
| | - Connie J Wang
- Department of Medicine, Division of Nephrology, The Kidney Institute
| | - Bertram L Kasiske
- Department of Medicine, Division of Nephrology, Hennepin County Medical Center, Minneapolis, Minnesota; and
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
221
|
Büscher R, Büscher AK, Weber S, Mohr J, Hegen B, Vester U, Hoyer PF. Clinical manifestations of autosomal recessive polycystic kidney disease (ARPKD): kidney-related and non-kidney-related phenotypes. Pediatr Nephrol 2014; 29:1915-25. [PMID: 24114580 DOI: 10.1007/s00467-013-2634-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Revised: 09/05/2013] [Accepted: 09/06/2013] [Indexed: 02/23/2023]
Abstract
Autosomal recessive polycystic kidney disease (ARPKD), although less frequent than the dominant form, is a common, inherited ciliopathy of childhood that is caused by mutations in the PKHD1-gene on chromosome 6. The characteristic dilatation of the renal collecting ducts starts in utero and can present at any stage from infancy to adulthood. Renal insufficiency may already begin in utero and may lead to early abortion or oligohydramnios and lung hypoplasia in the newborn. However, there are also affected children who have no evidence of renal dysfunction in utero and who are born with normal renal function. Up to 30 % of patients die in the perinatal period, and those surviving the neonatal period reach end stage renal disease (ESRD) in infancy, early childhood or adolescence. In contrast, some affected patients have been diagnosed as adults with renal function ranging from normal to moderate renal insufficiency to ESRD. The clinical spectrum of ARPKD is broader than previously recognized. While bilateral renal enlargement with microcystic dilatation is the predominant clinical feature, arterial hypertension, intrahepatic biliary dysgenesis remain important manifestations that affect approximately 45 % of infants. All patients with ARPKD develop clinical findings of congenital hepatic fibrosis (CHF); however, non-obstructive dilation of the intrahepatic bile ducts in the liver (Caroli's disease) is seen at the histological level in only a subset of patients. Cholangitis and variceal bleeding, sequelae of portal hypertension, are life-threatening complications that may occur more often in advanced cases of liver disease. In this review we focus on common and uncommon kidney-related and non-kidney-related phenotypes. Clinical management of ARPKD patients should include consideration of potential problems related to these manifestations.
Collapse
Affiliation(s)
- Rainer Büscher
- Children's Hospital, Pediatrics II, University of Duisburg-Essen, Hufelandstr. 55, 45122, Essen, Germany,
| | | | | | | | | | | | | |
Collapse
|
222
|
Sussman CR, Ward CJ, Leightner AC, Smith JL, Agarwal R, Harris PC, Torres VE. Phosphodiesterase 1A modulates cystogenesis in zebrafish. J Am Soc Nephrol 2014; 25:2222-30. [PMID: 24700876 PMCID: PMC4178429 DOI: 10.1681/asn.2013040421] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Accepted: 01/21/2014] [Indexed: 12/17/2022] Open
Abstract
Substantial evidence indicates the importance of elevated cAMP in polycystic kidney disease (PKD). Accumulation of cAMP in cystic tissues may be, in part, caused by enhanced adenylyl cyclase activity, but inhibition of cAMP degradation by phosphodiesterases (PDE) likely has an important role, because cAMP is inactivated much faster than it is synthesized. PDE1 is the only PDE family activated by Ca(2+), which is reduced in PKD cells. To assess the contribution of the PDE1A subfamily to renal cyst formation, we examined the expression and function of PDE1A in zebrafish. We identified two splice isoforms with alternative starts corresponding to human PDE1A1 and PDE1A4. Expression of the two isoforms varied in embryos and adult tissues, and both isoforms hydrolyzed cAMP with Ca(2+)/calmodulin dependence. Depletion of PDE1A in zebrafish embryos using splice- and translation-blocking morpholinos (MOs) caused pronephric cysts, hydrocephalus, and body curvature. Human PDE1A RNA and the PKA inhibitors, H89 and Rp-cAMPS, partially rescued phenotypes of pde1a morphants. Additionally, MO depletion of PDE1A aggravated phenotypes in pkd2 morphants, causing more severe body curvature, and human PDE1A RNA partially rescued pkd2 morphant phenotypes, pronephric cysts, hydrocephalus, and body curvature. Together, these data indicate the integral role of PDE1A and cAMP signaling in renal development and cystogenesis, imply that PDE1A activity is altered downstream of polycystin-2, and suggest that PDE1A is a viable drug target for PKD.
Collapse
Affiliation(s)
- Caroline R Sussman
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Christopher J Ward
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Amanda C Leightner
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jordan L Smith
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Reema Agarwal
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Peter C Harris
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Vicente E Torres
- Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, Minnesota
| |
Collapse
|
223
|
Kurschat CE, Müller RU, Franke M, Maintz D, Schermer B, Benzing T. An approach to cystic kidney diseases: the clinician's view. Nat Rev Nephrol 2014; 10:687-99. [DOI: 10.1038/nrneph.2014.173] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
224
|
Kang YR, Ahn JH, Kim KH, Choi YM, Choi J, Park JR. Multiple cardiovascular manifestations in a patient with autosomal dominant polycystic kidney disease. J Cardiovasc Ultrasound 2014; 22:144-7. [PMID: 25309692 PMCID: PMC4192413 DOI: 10.4250/jcu.2014.22.3.144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/27/2014] [Accepted: 08/20/2014] [Indexed: 01/13/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a systemic disorder associated with various extrarenal complications. The major cardiovascular complications of ADPKD include valvulopathies and vascular ectasia. A 64-year-old man who was diagnosed with ADPKD seven years previously was admitted to our hospital for heart failure. Pelvic computed tomography revealed multiple variable-sized cysts in both kidneys. Transthoracic echocardiography showed enlargement of the left ventricle and left atrium. Severe mitral regurgitation and moderate aortic regurgitation with annuloaortic ectasia were observed. The left main coronary artery was dilated. The patient had various cardiovascular features associated with ADPKD.
Collapse
Affiliation(s)
- Young Ran Kang
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jong-Hwa Ahn
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Kye Hwan Kim
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Young Min Choi
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jungwoo Choi
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| | - Jeong Rang Park
- Department of Internal Medicine, Gyeongsang National University Hospital, Gyeongsang National University School of Medicine, Jinju, Korea
| |
Collapse
|
225
|
Hoshino J, Suwabe T, Hayami N, Sumida K, Mise K, Kawada M, Imafuku A, Hiramatsu R, Yamanouchi M, Hasegawa E, Sawa N, Takei R, Takaichi K, Ubara Y. Survival after arterial embolization therapy in patients with polycystic kidney and liver disease. J Nephrol 2014; 28:369-77. [PMID: 25230989 DOI: 10.1007/s40620-014-0138-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 09/01/2014] [Indexed: 02/07/2023]
Abstract
BACKGROUND Transcatheter arterial embolization (TAE) has become a therapeutic option for symptomatic polycystic kidney disease (PKD) and polycystic liver disease (PLD). However, factors affecting survival with renal TAE remain unknown. METHODS All symptomatic patients with severe PKD and/or PLD who received renal and/or hepatic TAE at our center from October 1996 through March 2013 (n = 1,028) were followed until death. Their survival was compared with that of the general PKD population on dialysis in Japan. Factors affecting survival were analyzed using the Cox hazard model. RESULTS After renal TAE, 5- and 10-year survival was, respectively, 0.78 (95% confidence interval, 0.74-0.82) and 0.56 (0.49-0.63); with hepatic TAE, 0.69 (0.58-0.77) and 0.41 (0.22-0.60); and with dual TAE (renal and hepatic), 0.82 (0.72-0.88) and 0.45 (0.31-0.59). Survival after dialysis initiation was better among patients with renal TAE than among general PKD patients. Factors affecting survival after renal TAE were age [hazard ratio (HR) 3.02 (1.44-6.33) for every 10 years] and albumin [HR 0.70 (0.55-0.89) per 0.1 g/dl]. Kidney volume was not associated with patient death after TAE. The main causes of death among patients after renal TAE were similar to those of the general PKD population on dialysis whereas, after hepatic TAE, the main cause was cyst infection with liver failure (12.5% with PLD and 5.9% with PKD, p < 0.01). CONCLUSION Survival after renal TAE with severe PKD was better than for the general PKD population on dialysis, suggesting that renal TAE could overcome the disadvantage due to huge organ size.
Collapse
Affiliation(s)
- Junichi Hoshino
- Nephrology Center, Toranomon Hospital, 1-3-1, Kajigaya, Takatsu-ku, Kawasaki, Kanagawa, 213-8587, Japan,
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Cristea O, Yanko D, Felbel S, House A, Sener A, Luke PPW. Maximal kidney length predicts need for native nephrectomy in ADPKD patients undergoing renal transplantation. Can Urol Assoc J 2014; 8:278-82. [PMID: 25210553 DOI: 10.5489/cuaj.2128] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
INTRODUCTION Native nephrectomy in patients with autosomal dominant polycystic kidney disease (ADPKD) is performed on a case-by-case basis. We determine if pre-transplant maximal kidney length (MKL) can be used to predict ultimate nephrectomy status. METHODS We performed a retrospective review of ADPKD patients who underwent renal transplantation at our centre between January 2000 and December 2012. Pre-transplant measurements of MKL alone, MKL adjusted for height (HtMKL), weight (WtMKL) and body mass index (BMI-MKL) were each assessed for their predictive ability via a receiver operating characteristic (ROC) curve analysis. RESULTS In total, 84 patients met our inclusion criteria, of which 17 (20.2%) underwent native nephrectomy. An MKL ROC curve analysis revealed an area under the curve (AUC) of 0.867 (95% confidence interval [CI] 0.775-0.931; p < 0.001). An optimal cutoff criterion of >21.5 cm revealed a sensitivity of 94.1% (95% CI 71.3-99.9) and specificity of 70.1% (95% CI 57.7-80.7) for eventual nephrectomy. The AUC of HtMKL, WtMKL and BMI-MKL ROC curves did not differ significantly from MKL alone. HtMKL improved specificity, but not overall test performance. The determination of the cut-off MKL may be influenced by the single-centre retrospective nature of this analysis, as well as the fact that renal size was determined by ultrasound and not computerized tomography or magnetic resonance imaging. CONCLUSION MKL in patients with ADPKD is associated with the eventual need for nephrectomy and may be a useful clinical tool to risk stratify these patients and therefore guide patient conversations to a decision to leave the native kidneys in situ.
Collapse
Affiliation(s)
- Octav Cristea
- Schulich School of Medicine & Dentistry, Western University, London, ON
| | - Daniel Yanko
- Department of Surgery, Division of Urology, Western University, London, ON
| | - Sarah Felbel
- Department of Surgery, Division of Urology, Western University, London, ON
| | - Andrew House
- Department of Medicine, Division of Nephrology, Western University, London, ON
| | - Alp Sener
- Schulich School of Medicine & Dentistry, Western University, London, ON; ; Department of Surgery, Division of Urology, Western University, London, ON; ; Multi-Organ Transplant Program, London Health Sciences Centre, Western University, London, ON; ; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, Western University, London, ON
| | - Patrick P W Luke
- Schulich School of Medicine & Dentistry, Western University, London, ON; ; Department of Surgery, Division of Urology, Western University, London, ON; ; Multi-Organ Transplant Program, London Health Sciences Centre, Western University, London, ON; ; Matthew Mailing Centre for Translational Transplant Studies, London Health Sciences Centre, Western University, London, ON
| |
Collapse
|
227
|
Paul BM, Vanden Heuvel GB. Kidney: polycystic kidney disease. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2014; 3:465-87. [PMID: 25186187 DOI: 10.1002/wdev.152] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Revised: 07/14/2014] [Accepted: 07/29/2014] [Indexed: 12/22/2022]
Abstract
Polycystic kidney disease (PKD) is a life-threatening genetic disorder characterized by the presence of fluid-filled cysts primarily in the kidneys. PKD can be inherited as autosomal recessive (ARPKD) or autosomal dominant (ADPKD) traits. Mutations in either the PKD1 or PKD2 genes, which encode polycystin 1 and polycystin 2, are the underlying cause of ADPKD. Progressive cyst formation and renal enlargement lead to renal insufficiency in these patients, which need to be managed by lifelong dialysis or renal transplantation. While characteristic features of PKD are abnormalities in epithelial cell proliferation, fluid secretion, extracellular matrix and differentiation, the molecular mechanisms underlying these events are not understood. Here we review the progress that has been made in defining the function of the polycystins, and how disruption of these functions may be involved in cystogenesis.
Collapse
Affiliation(s)
- Binu M Paul
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
228
|
ACG clinical guideline: the diagnosis and management of focal liver lesions. Am J Gastroenterol 2014; 109:1328-47; quiz 1348. [PMID: 25135008 DOI: 10.1038/ajg.2014.213] [Citation(s) in RCA: 256] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 05/22/2014] [Indexed: 12/11/2022]
Abstract
Focal liver lesions (FLL) have been a common reason for consultation faced by gastroenterologists and hepatologists. The increasing and widespread use of imaging studies has led to an increase in detection of incidental FLL. It is important to consider not only malignant liver lesions, but also benign solid and cystic liver lesions such as hemangioma, focal nodular hyperplasia, hepatocellular adenoma, and hepatic cysts, in the differential diagnosis. In this ACG practice guideline, the authors provide an evidence-based approach to the diagnosis and management of FLL.
Collapse
|
229
|
Williamson A, Paterson S, Erolin C, Sweeney C, Townell N, Nabi G. Laparoscopic nephrectomy for adult polycystic kidney disease: safety, feasibility, and early outcomes. J Endourol 2014; 28:1268-77. [PMID: 24999965 DOI: 10.1089/end.2014.0311] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND AND PURPOSE Indications for laparoscopic renal surgery are increasing; however, benefits in adult polycystic kidney disease (APKD) remain uncertain. Our objective was to systematically synthesize the reported literature on safety, feasibility, complications, and early outcomes of laparoscopic nephrectomy in APKD to determine clinical benefits for surgical practice. METHODS We conducted a meta-analysis of the published literature reporting on laparoscopic nephrectomy in APKD between 1991 and 2013. The criteria from the Strengthening the Reporting of Observational Studies in Epidemiology (STROBE) study were used to assess the quality of reported literature. RESULTS One prospective and 15 retrospective studies of low to modest quality (according to the STROBE checklist) were identified, reporting on 293 patients who underwent laparoscopic nephrectomy for APKD. None of the studies was a randomized clinical trial (RCT). The transperitoneal approach was the most commonly used technique. Body mass index ranged from 16 to 57 (mean 26.2 kg/m(2); 53% of patients were dialysis dependent, and 31% had a previous or simultaneous transplant. Kidney length ranged from 8 to 50 cm (mean 34.5cm), and the mean mass of affected kidneys was 1647 g (range 132 g-7200 g). Duration of hospital stay ranged from 2.6 to 11 days (mean 4.9 days). Operative time ranged from 90 to 568 minutes, with 16.2% of patients needing blood transfusion. There were 24 intraoperative complications and 68 postoperative complications, a rate of 8% and 24%, respectively. A total of 16 (5%) cases were converted to an open technique. No mortality was reported in any of the included studies. CONCLUSION The quality of the included studies is poor, and it is difficult to argue for or against change in clinical practice because the evidence included is of level 3 and 4 only. Higher quality studies are needed to demonstrate that the technique is generalizable across all populations.
Collapse
Affiliation(s)
- Adam Williamson
- Academic Department of Urology, Ninewells Hospital and Medical School , Dundee, Scotland, United Kingdom
| | | | | | | | | | | |
Collapse
|
230
|
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 or PKD2, which encode polycystin-1 and polycystin-2, respectively. Rodent models are available to study the pathogenesis of polycystic kidney disease (PKD) and for preclinical testing of potential therapies-either genetically engineered models carrying mutations in Pkd1 or Pkd2 or models of renal cystic disease that do not have mutations in these genes. The models are characterized by age at onset of disease, rate of disease progression, the affected nephron segment, the number of affected nephrons, synchronized or unsynchronized cyst formation and the extent of fibrosis and inflammation. Mouse models have provided valuable mechanistic insights into the pathogenesis of PKD; for example, mutated Pkd1 or Pkd2 cause renal cysts but additional factors are also required, and the rate of cyst formation is increased in the presence of renal injury. Animal studies have also revealed complex genetic and functional interactions among various genes and proteins associated with PKD. Here, we provide an update on the preclinical models commonly used to study the molecular pathogenesis of ADPKD and test potential therapeutic strategies. Progress made in understanding the pathophysiology of human ADPKD through these animal models is also discussed.
Collapse
Affiliation(s)
- Hester Happé
- Department of Human Genetics, Leiden University Medical Center, S4-P, PO Box 9600, Albinusdreef 2, Leiden, 2333 ZA Leiden, Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, S4-P, PO Box 9600, Albinusdreef 2, Leiden, 2333 ZA Leiden, Netherlands
| |
Collapse
|
231
|
Abstract
Mutations inactivating the cilia-localized Pkd1 protein result in autosomal dominant polycystic kidney disease (ADPKD), a serious inherited syndrome affecting ∼ 1 in 500 people, in which accumulation of renal cysts eventually destroys kidney function. Severity of ADPKD varies throughout the population, for reasons thought to involve differences both in intragenic Pkd1 mutations and in modifier alleles. The scaffolding protein NEDD9, commonly dysregulated during cancer progression, interacts with Aurora-A (AURKA) kinase to control ciliary resorption, and with Src and other partners to influence proliferative signaling pathways often activated in ADPKD. We here demonstrate Nedd9 expression is deregulated in human ADPKD and a mouse ADPKD model. Although genetic ablation of Nedd9 does not independently influence cystogenesis, constitutive absence of Nedd9 strongly promotes cyst formation in the tamoxifen-inducible Pkd1fl/fl;Cre/Esr1(+) mouse model of ADPKD. This cystogenic effect is associated with striking morphological defects in the cilia of Pkd1(-/-);Nedd9(-/-) mice, associated with specific loss of ciliary localization of adenylase cyclase III in the doubly mutant genotype. Ciliary phenotypes imply a failure of Aurora-A activation: Compatible with this idea, Pkd1(-/-);Nedd9(-/-) mice had ciliary resorption defects, and treatment of Pkd1(-/-) mice with a clinical Aurora-A kinase inhibitor exacerbated cystogenesis. In addition, activation of the ADPKD-associated signaling effectors Src, Erk, and the mTOR effector S6 was enhanced, and Ca(2+) response to external stimuli was reduced, in Pkd1(-/-);Nedd9(-/-) versus Pkd1(-/-) mice. Together, these results indicated an important modifier action of Nedd9 on ADPKD pathogenesis involving failure to activate Aurora-A.
Collapse
|
232
|
Wisenbaugh ES, Tyson MD, Castle EP, Humphreys MR, Andrews PE. Massive renal size is not a contraindication to a laparoscopic approach for bilateral native nephrectomies in autosomal dominant polycystic kidney disease (ADPKD). BJU Int 2014; 115:796-801. [DOI: 10.1111/bju.12821] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | - Mark D. Tyson
- Department of Urology; Mayo Clinic Hospital; Phoenix AZ USA
| | - Erik P. Castle
- Department of Urology; Mayo Clinic Hospital; Phoenix AZ USA
| | | | | |
Collapse
|
233
|
Ferraz RRN, Fonseca JM, Germino GG, Onuchic LF, Heilberg IP. Determination of urinary lithogenic parameters in murine models orthologous to autosomal dominant polycystic kidney disease. Urolithiasis 2014; 42:301-7. [PMID: 24817661 PMCID: PMC5602548 DOI: 10.1007/s00240-014-0664-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 04/15/2014] [Indexed: 10/25/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), a genetic disease caused by mutations in PKD1 or PKD2 genes, is associated with a high prevalence of nephrolithiasis. The underlying mechanisms may encompass structural abnormalities resulting from cyst growth, urinary metabolic abnormalities or both. An increased frequency of hypocitraturia has been described in ADPKD even in the absence of nephrolithiasis, suggesting that metabolic alterations may be associated with ADPKD per se. We aimed to investigate whether non-cystic Pkd1-haploinsufficient (Pkd1(+/-)) and/or nestin-Cre Pkd1-targeted cystic (Pkd1(cond/cond):Nestin(cre)) mouse models develop urinary metabolic abnormalities potentially related to nephrolithiasis in ADPKD. 24-h urine samples were collected during three non-consecutive days from 10-12 and 18-20 week-old animals. At 10-12 weeks of age, urinary oxalate, calcium, magnesium, citrate and uric acid did not differ between test and their respective control groups. At 18-20 weeks, Pkd1(+/-) showed slightly but significantly higher urinary uric acid vs. controls while cystic animals did not. The absence of hypocitraturia, hyperoxaluria and hyperuricosuria in the cystic model at both ages and the finding of hyperuricosuria in the 18-20 week-old animals suggest that anatomic cystic distortions per se do not generate the metabolic disturbances described in human ADPKD-related nephrolithiasis, while Pkd1 haploinsufficiency may contribute to this phenotype in this animal model.
Collapse
Affiliation(s)
| | | | | | - Luiz Fernando Onuchic
- Division of Nephrology, University of Sao Paulo School of Medicine, Sao Paulo, Brazil
| | | |
Collapse
|
234
|
Reinecke NL, Cunha TM, Heilberg IP, Higa EMS, Nishiura JL, Neder JA, Almeida WS, Schor N. Exercise Capacity in Polycystic Kidney Disease. Am J Kidney Dis 2014; 64:239-46. [DOI: 10.1053/j.ajkd.2014.03.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 03/18/2014] [Indexed: 11/11/2022]
|
235
|
Blanchette CM, Iorga ŞR, Altan A, Seare JG, Fan Y, Rossetti S, Gutierrez B. Healthcare Resource Utilization and Costs Associated with Autosomal Dominant Polycystic Kidney Disease. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2014; 2:63-74. [PMID: 37664083 PMCID: PMC10471403 DOI: 10.36469/9889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Background: Autosomal dominant polycystic kidney disease (ADPKD), a hereditary nephropathy, eventually leads to end-stage renal disease (ESRD), typically by mid-life. Objectives: The objective of this study was to assess real-world healthcare resource utilization and cost among commercially insured (COM) and Medicare Advantage (MAPD) ADPKD patients in addition to the cost profile by chronic kidney disease (CKD) stage. Methods: Patients diagnosed with ADPKD (two or more claims) with ≥30 days of continuous medical and pharmacy benefits and no evidence of autosomal recessive polycystic kidney disease were selected (Optum Research Database and Impact National Benchmarking Database: 1/1/06-8/31/12). Plan and patient paid healthcare costs and resource utilization per patient per month (PPPM) were described in total and by insurance type. CKD stage was established based on serum creatinine laboratory values or dialysis-related codes. Adjusted, CKD stage-specific costs were predicted for 4 years using regression models. Results: Of the 36,253,096 patients in the databases (1/1/06-8/31/12), 5,051 had evidence of ADPKD. Following exclusion criteria, 4,356 COM and 468 MAPD ADPKD patients remained. Total healthcare resource utilization and costs were high, and costs increased substantially from CKD stage 1-5. PPPM healthcare costs were 37% for ADPKD management and 52% for dialysis services. Predicted 4-year healthcare costs by CKD stage were $40,164 (stage 1), $33,397 (stage 2), $42,686 (stage 3), $148,402 (stage 4), and $207,548 (stage 5). Conclusions: Healthcare resource utilization and costs associated with ADPKD were substantial, irrespective of payer type, and primarily driven by CKD stage. Of the total healthcare costs, 88% were ADPKD- and dialysis-related. Most impactful was the spike in predicted cost when patients progressed from CKD stage 3 to stage 4 (by 348%) after multivariate adjustment. These stage 4-associated costs are primarily due to ultimate progression into stage 5 and ESRD within the 4-year time frame.
Collapse
Affiliation(s)
- Christopher M Blanchette
- Otsuka America Pharmaceutical, Inc., Princeton, NJ, USA; University of North Carolina, Charlotte, NC, USA
| | | | | | | | - Ying Fan
- OptumInsight, Eden Prairie, MN, USA
| | | | | |
Collapse
|
236
|
Mallett A, Patel C, Salisbury A, Wang Z, Healy H, Hoy W. The prevalence and epidemiology of genetic renal disease amongst adults with chronic kidney disease in Australia. Orphanet J Rare Dis 2014; 9:98. [PMID: 24980890 PMCID: PMC4085397 DOI: 10.1186/1750-1172-9-98] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2014] [Accepted: 06/10/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There are an established and growing number of Mendelian genetic causes for chronic kidney disease (CKD) in adults, though estimates of prevalence have been speculative. The CKD Queensland (CKD.QLD) registry enables partial clarification of this through the study of adults with CKD receiving nephrology care throughout Queensland, Australia. METHODS Data from the first 2,935 patients consented to the CKD.QLD registry across five sites was analysed, with a comparison between those with and without Genetic Renal Disease (GRD). Prevalence of GRD amongst those with diagnosed CKD, the general population, and commencing renal replacement therapy (RRT) was calculated using the CKD.QLD registry, national census data and extracted Australian and New Zealand Dialysis and Transplantation (ANZDATA) registry report data respectively. RESULTS Patients with GRD constituted 9.8% of this Australian adult CKD cohort (287/2935). This was lower than in local incident RRT cohorts (2006-2011: 9.8% vs 11.3%, x2 = 0.014). Cases of adult CKD GRD were more likely to be female (54.0% vs 45.6%; x2 = 0.007), younger (mean 52.6 yrs vs 69.3 yrs, p < 0.001), have a higher eGFR (mean 49.7 ml/min/1.73 m2 vs 40.4 ml/min/1.73 m2, p < 0.001), and have earlier stage renal disease (CKD Stage 1: 15.7% vs 5.1%, x2 < 0.0005) than those without GRD. CONCLUSIONS The proportion of GRD amongst an Australian adult CKD population in specialty renal practice is similar to past estimations. GRD is a significant cause for CKD and for RRT commencement, presenting opportunities for ongoing longitudinal study, directed therapeutics and clinical service redesign.
Collapse
Affiliation(s)
- Andrew Mallett
- Department of Renal Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia.
| | | | | | | | | | | |
Collapse
|
237
|
Kanaan N, Devuyst O, Pirson Y. Renal transplantation in autosomal dominant polycystic kidney disease. Nat Rev Nephrol 2014; 10:455-65. [PMID: 24935705 DOI: 10.1038/nrneph.2014.104] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In patients with autosomal dominant polycystic kidney disease (ADPKD) evaluated for kidney transplantation, issues related to native nephrectomy, cystic liver involvement, screening for intracranial aneurysms and living-related kidney donation deserve special consideration. Prophylactic native nephrectomy is restricted to patients with a history of cyst infection or recurrent haemorrhage or to those in whom space must be made to implant the graft. Patients with liver involvement require pretransplant imaging. Selection of patients for pretransplant screening of intracranial aneurysms should follow the general recommendations for patients with ADPKD. In living related-donor candidates aged <30 years and at-risk of ADPKD, molecular genetic testing should be carried out when ultrasonography and MRI findings are normal or equivocal. After kidney transplantation, patient and graft survival rates are excellent and the volume of native kidneys decreases. However, liver cysts continue to grow and treatment with a somatostatin analogue should be considered in patients with massive cyst involvement. Cerebrovascular events have a marginal effect on post-transplant morbidity and mortality. An increased risk of new-onset diabetes mellitus and nonmelanoma skin cancers has been reported, but several studies have challenged these findings. Finally, no data currently support the preferential use of mammalian target of rapamycin inhibitors as immunosuppressive agents in transplant recipients with ADPKD.
Collapse
Affiliation(s)
- Nada Kanaan
- Division of Nephrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Olivier Devuyst
- Division of Nephrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Yves Pirson
- Division of Nephrology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 Avenue Hippocrate, B-1200 Brussels, Belgium
| |
Collapse
|
238
|
Chang MY, Ong ACM. New treatments for autosomal dominant polycystic kidney disease. Br J Clin Pharmacol 2014; 76:524-35. [PMID: 23594398 DOI: 10.1111/bcp.12136] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 03/21/2013] [Indexed: 02/06/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease and results from mutations in PKD1 or PKD2. Cyst initiation and expansion arise from a combination of abnormal cell proliferation, fluid secretion and extracellular matrix defects and results in kidney enlargement and interstitial fibrosis. Since its first description over 200 years ago, ADPKD has been considered an untreatable condition and its management is limited to blood pressure reduction and symptomatic treatment of disease complications. Results of the recently reported TEMPO 3/4 trial thus represent a paradigm shift in demonstrating for the first time that cystic disease and loss of renal function can be slowed in humans. In this paper, we review the major therapeutic strategies currently being explored in ADPKD including a range of novel approaches in preclinical models. It is anticipated that the clinical management of ADPKD will undergo a revolution in the next decade with the translation of new treatments into routine clinical use.
Collapse
Affiliation(s)
- Ming-Yang Chang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | | |
Collapse
|
239
|
Braun WE, Schold JD, Stephany BR, Spirko RA, Herts BR. Low-dose rapamycin (sirolimus) effects in autosomal dominant polycystic kidney disease: an open-label randomized controlled pilot study. Clin J Am Soc Nephrol 2014; 9:881-8. [PMID: 24721888 PMCID: PMC4011437 DOI: 10.2215/cjn.02650313] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 01/04/2014] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVES The two largest studies of mammalian target of rapamycin inhibitor treatment of autosomal dominant polycystic kidney disease (ADPKD) demonstrated no clear benefit on the primary endpoint of total kidney volume (TKV) or on eGFR. The present study evaluated two levels of rapamycin on the 12-month change in (125)I-iothalamate GFR (iGFR) as the primary endpoint and TKV secondarily. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS In a 12-month open-label pilot study, 30 adult patients with ADPKD were randomly assigned to low-dose (LD) rapamycin (rapamycin trough blood level, 2-5 ng/ml) (LD group, n=10), standard-dose (STD) rapamycin trough level (>5-8 ng/ml) (STD group, n=10), or standard care (SC group, n=10). They were evaluated with iGFR and noncontrast computed tomography. RESULTS Change in iGFR at 12 months was significantly higher in the LD group (7.7±12.5 ml/min per 1.73 m(2); n=9) than in the SC group (-11.2 ± 9.1 ml/min per 1.73 m(2); n=9) (LD versus SC: P<0.01). Change in iGFR at 12 months in the STD group (1.6 ± 12.1 ml/min per 1.73 m(2); n=8) was not significantly greater than that in the SC group (P=0.07), but it was in the combined treatment groups (LD+STD versus SC: P<0.01). Neither eGFR calculated by the CKD-Epidemiology Collaboration equation nor TKV (secondary endpoint) changed significantly from baseline to 12 months in any of the groups. On the basis of results of the mixed model, during the study, patients in the LD group had significantly lower trough blood levels of rapamycin (mean range ± SD, 2.40 ± 0.64 to 2.90 ± 1.20 ng/ml) compared with those in the STD group (3.93 ± 2.27 to 5.77 ± 1.06 ng/ml) (P<0.01). CONCLUSION Patients with ADPKD receiving LD rapamycin demonstrated a significant increase in iGFR compared with those receiving standard care, without a significant effect on TKV after 12 months.
Collapse
Affiliation(s)
- William E Braun
- Glickman Urological and Kidney Institute,, †Quantitative Health Sciences, and, ‡Imaging Institute, Cleveland Clinic, Cleveland, Ohio
| | | | | | | | | |
Collapse
|
240
|
Shaw C, Simms RJ, Pitcher D, Sandford R. Epidemiology of patients in England and Wales with autosomal dominant polycystic kidney disease and end-stage renal failure. Nephrol Dial Transplant 2014; 29:1910-8. [DOI: 10.1093/ndt/gfu087] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
241
|
Cnossen WR, te Morsche RHM, Hoischen A, Gilissen C, Chrispijn M, Venselaar H, Mehdi S, Bergmann C, Veltman JA, Drenth JPH. Whole-exome sequencing reveals LRP5 mutations and canonical Wnt signaling associated with hepatic cystogenesis. Proc Natl Acad Sci U S A 2014; 111:5343-8. [PMID: 24706814 PMCID: PMC3986119 DOI: 10.1073/pnas.1309438111] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Polycystic livers are seen in the rare inherited disorder isolated polycystic liver disease (PCLD) and are recognized as the most common extrarenal manifestation in autosomal dominant polycystic kidney disease. Hepatic cystogenesis is characterized by progressive proliferation of cholangiocytes, ultimately causing hepatomegaly. Genetically, polycystic liver disease is a heterogeneous disorder with incomplete penetrance and caused by mutations in PRKCSH, SEC63, PKD1, or PKD2. Genome-wide SNP typing and Sanger sequencing revealed no pathogenic variants in hitherto genes in an extended PCLD family. We performed whole-exome sequencing of DNA samples from two members. A heterozygous variant c.3562C > T located at a highly conserved amino acid position (p.R1188W) in the low density lipoprotein receptor-related protein 5 (LRP5) gene segregated with the disease (logarithm of odds score, 4.62) but was not observed in more than 1,000 unaffected individuals. Screening of LRP5 in a PCLD cohort identified three additional mutations in three unrelated families with polycystic livers (p.V454M, p.R1529S, and p.D1551N), again all undetected in controls. All variants were predicted to be damaging with profound structural effects on LRP5 protein domains. Liver cyst tissue and normal hepatic tissue samples from patients and controls showed abundant LRP5 expression by immunohistochemistry. Functional activity analyses indicated that mutant LRP5 led to reduced wingless signal activation. In conclusion, we demonstrate that germ-line LRP5 missense mutations are associated with hepatic cystogenesis. The findings presented in this study link the pathophysiology of PCLD to deregulation of the canonical wingless signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | | | - Hanka Venselaar
- Center for Molecular and Biomolecular Informatics, Institute for Genetic and Metabolic Disease, Radboud university medical center, 6500 HB Nijmegen, The Netherlands
| | - Soufi Mehdi
- Department of Digestive and Oncological Surgery, Faculty of Medicine, University Mohammed First, 60000 Oujda, Morocco
| | - Carsten Bergmann
- Center for Human Genetics, Bioscientia, 55218 Ingelheim, Germany; and
- Department of Nephrology and Center for Clinical Research, University Hospital Freiburg, 79106 Freiburg, Germany
| | | | | |
Collapse
|
242
|
Baur BP, Meaney CJ. Review of tolvaptan for autosomal dominant polycystic kidney disease. Pharmacotherapy 2014; 34:605-16. [PMID: 24706579 DOI: 10.1002/phar.1421] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by bilateral renal cysts, kidney pain, hypertension, and progressive loss of renal function. It is a leading cause of end-stage renal disease and the most common inherited kidney disease in the United States. Despite its prevalence, disease-modifying treatment options do not currently exist. Tolvaptan is an orally active, selective arginine vasopressin V2 receptor antagonist already in use for hyponatremia. Tolvaptan exhibits dose-proportional pharmacokinetics with a half-life of ~12 hours. Metabolism occurs through the cytochrome P450 3A4 isoenzyme, and tolvaptan is a substrate for P-glycoprotein, resulting in numerous drug interactions. Recent research has highlighted the beneficial effect of tolvaptan on delaying the progression of ADPKD, which is the focus of this review. Pharmacologic, preclinical, and phase II and III clinical trial studies have demonstrated that tolvaptan is an effective treatment option that targets underlying pathogenic mechanisms of ADPKD. Tolvaptan delays the increase in total kidney volume (surrogate marker for disease progression), slows the decline in renal function, and reduces kidney pain. However, tolvaptan has significant adverse effects including aquaretic effects (polyuria, nocturia, polydipsia) and elevation of aminotransferase enzyme concentrations with the potential for acute liver failure. Appropriate patient selection is critical to optimize long-term benefits while minimizing adverse effects and hepatotoxic risk factors. Overall, tolvaptan is the first pharmacotherapeutic intervention to demonstrate significant benefit in the treatment of ADPKD, but practitioners and regulatory agencies must carefully weigh the risks versus benefits. Additional research should focus on incidence and risk factors of liver injury, cost-effectiveness, clinical management of drug-drug interactions, and long-term disease outcomes.
Collapse
Affiliation(s)
- Brian P Baur
- University at Buffalo School of Pharmacy and Pharmaceutical Sciences, Buffalo, New York
| | | |
Collapse
|
243
|
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: 1.9] [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
|
244
|
DesRochers TM, Palma E, Kaplan DL. Tissue-engineered kidney disease models. Adv Drug Deliv Rev 2014; 69-70:67-80. [PMID: 24361391 DOI: 10.1016/j.addr.2013.12.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 02/08/2023]
Abstract
Renal disease represents a major health problem that often results in end-stage renal failure necessitating dialysis and eventually transplantation. Historically these diseases have been studied with patient observation and screening, animal models, and two-dimensional cell culture. In this review, we focus on recent advances in tissue engineered kidney disease models that have the capacity to compensate for the limitations of traditional modalities. The cells and materials utilized to develop these models are discussed and tissue engineered models of polycystic kidney disease, drug-induced nephrotoxicity, and the glomerulus are examined in detail. The application of these models has the potential to direct future disease treatments and preclinical drug development.
Collapse
|
245
|
Abstract
INTRODUCTION Autosomal-dominant polycystic kidney disease (ADPKD) represents a therapeutic challenge as effective treatment to retard the growth of cysts in the kidneys and the liver has not been available despite decades of intense basic and clinical research. AREAS COVERED Several clinical trials have been performed in recent years to study the effect of diverse drugs on the growth of renal and hepatic cysts, and on functional deterioration of the glomerular filtration rate. The drug classes that have been tested in randomized clinical trials include the mammalian target of rapamycin (mTOR) inhibitors, sirolimus and everolimus, the somatostatin analogues (octreotide, lanreotide, pasireotide), and most recently, the vasopressin V2 receptor antagonist, tolvaptan. The results with the mTOR inhibitors were disappointing, but more encouraging with the somatostatin analogues and with tolvaptan. Additional drugs are being tested, which include among others, the SRC-ABL tyrosine kinase inhibitor, bosutinib, and the traditional Chinese herbal medication, triptolide. Additional therapeutic strategies to retard cyst growth aim at blood pressure control via inhibition of the renin-angiotensin system and the sympathetic nervous system. EXPERT OPINION Given the accumulated knowledge, it is currently uncertain whether drugs will become available in the near future to significantly change the course of the relentlessly progressing polycystic kidney disease.
Collapse
Affiliation(s)
- Rudolf P Wüthrich
- University Hospital, Division of Nephrology , Rämistrasse 100, 8091 Zürich , Switzerland +41 44 255 33 84 ; +41 44 255 45 93 ;
| | | |
Collapse
|
246
|
Vijayakumar S, Dang S, Marinkovich MP, Lazarova Z, Yoder B, Torres VE, Wallace DP. Aberrant expression of laminin-332 promotes cell proliferation and cyst growth in ARPKD. Am J Physiol Renal Physiol 2014; 306:F640-54. [PMID: 24370592 PMCID: PMC3949036 DOI: 10.1152/ajprenal.00104.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 12/23/2013] [Indexed: 11/22/2022] Open
Abstract
Basement membrane abnormalities have often been observed in kidney cysts of polycystic kidney disease (PKD) patients and animal models. There is an abnormal deposition of extracellular matrix molecules, including laminin-α3,β3,γ2 (laminin-332), in human autosomal dominant PKD (ADPKD). Knockdown of PKD1 paralogs in zebrafish leads to dysregulated synthesis of the extracellular matrix, suggesting that altered basement membrane assembly may be a primary defect in ADPKD. In this study, we demonstrate that laminin-332 is aberrantly expressed in cysts and precystic tubules of human autosomal recessive PKD (ARPKD) kidneys as well as in the kidneys of PCK rats, an orthologous ARPKD model. There was aberrant expression of laminin-γ2 as early as postnatal day 2 and elevated laminin-332 protein in postnatal day 30, coinciding with the formation and early growth of renal cysts in PCK rat kidneys. We also show that a kidney cell line derived from Oak Ridge polycystic kidney mice, another model of ARPKD, exhibited abnormal lumen-deficient and multilumen structures in Matrigel culture. These cells had increased proliferation rates and altered expression levels of laminin-332 compared with their rescued counterparts. A function-blocking polyclonal antibody to laminin-332 significantly inhibited their abnormal proliferation rates and rescued their aberrant phenotype in Matrigel culture. Furthermore, abnormal laminin-332 expression in cysts originating from collecting ducts and proximal tubules as well as in precystic tubules was observed in a human end-stage ADPKD kidney. Our results suggest that abnormal expression of laminin-332 contributes to the aberrant proliferation of cyst epithelial cells and cyst growth in genetic forms of PKD.
Collapse
Affiliation(s)
- Soundarapandian Vijayakumar
- Dept. of Natural Sciences and Mathematics, SUNY Cobleskill, 111 Schenectady Ave. WH200, Cobleskill, NY 12043.
| | | | | | | | | | | | | |
Collapse
|
247
|
Lian P, Li A, Li Y, Liu H, Liang D, Hu B, Lin D, Jiang T, Moeckel G, Qin D, Wu G. Loss of polycystin-1 inhibits Bicc1 expression during mouse development. PLoS One 2014; 9:e88816. [PMID: 24594709 PMCID: PMC3940423 DOI: 10.1371/journal.pone.0088816] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Accepted: 01/16/2014] [Indexed: 12/21/2022] Open
Abstract
Bicc1 is a mouse homologue of Drosophila Bicaudal-C (dBic-C), which encodes an RNA-binding protein. Orthologs of dBic-C have been identified in many species, from C. elegans to humans. Bicc1-mutant mice exhibit a cystic phenotype in the kidney that is very similar to human polycystic kidney disease. Even though many studies have explored the gene characteristics and its functions in multiple species, the developmental profile of the Bicc1 gene product (Bicc1) in mammal has not yet been completely characterized. To this end, we generated a polyclonal antibody against Bicc1 and examined its spatial and temporal expression patterns during mouse embryogenesis and organogenesis. Our results demonstrated that Bicc1 starts to be expressed in the neural tube as early as embryonic day (E) 8.5 and is widely expressed in epithelial derivatives including the gut and hepatic cells at E10.5, and the pulmonary bronchi at E11.5. In mouse kidney development, Bicc1 appears in the early ureteric bud and mesonephric tubules at E11.5 and is also expressed in the metanephros at the same stage. During postnatal kidney development, Bicc1 expression gradually expands from the cortical to the medullary and papillary regions, and it is highly expressed in the proximal tubules. In addition, we discovered that loss of the Pkd1 gene product, polycystin-1 (PC1), whose mutation causes human autosomal dominant polycystic kidney disease (ADPKD), downregulates Bicc1 expression in vitro and in vivo. Our findings demonstrate that Bicc1 is developmentally regulated and reveal a new molecular link between Bicc1 and Pkd1.
Collapse
Affiliation(s)
- Peiwen Lian
- Division of Translational Cancer Research and Therapy, State Key Laboratory of Molecular Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Ao Li
- Division of Translational Cancer Research and Therapy, State Key Laboratory of Molecular Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Yuan Li
- Division of Translational Cancer Research and Therapy, State Key Laboratory of Molecular Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haichao Liu
- Division of Translational Cancer Research and Therapy, State Key Laboratory of Molecular Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Dan Liang
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Bo Hu
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
| | - De Lin
- Department of Biochemistry, Vanderbilt University, Nashville, Tennessee, United States of America
| | - Tang Jiang
- Department of Medicine, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Dahui Qin
- Department of Pathology, Moffitt Cancer Center, Tampa, Florida, United States of America
| | - Guanqing Wu
- Division of Translational Cancer Research and Therapy, State Key Laboratory of Molecular Oncology, Cancer Hospital and Institute, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, United States of America
- Department of Cell & Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States of America
| |
Collapse
|
248
|
Raptis V, Georgianos PI, Sarafidis PA, Sioulis A, Makedou K, Makedou A, Grekas DM, Kapoulas S. Elevated asymmetric dimethylarginine is associated with oxidant stress aggravation in patients with early stage autosomal dominant polycystic kidney disease. Kidney Blood Press Res 2014; 38:72-82. [PMID: 24577239 DOI: 10.1159/000355756] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND/AIMS In experimental models of polycystic kidney disease impaired bioavailability of nitric oxide (NO) and elevated mRNA expression of oxidative stress markers at the kidney level was noted. However, clinical studies investigating the potential role of endothelial dysfunction and oxidative stress in the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD) are limited. We evaluated asymmetric dimethylarginine (ADMA) as marker of NO synthase inhibitor as well as 15-F2t-Isoprostane and oxidized-low density lipoprotein (oxidized-LDL) as measures of oxidative stress in patients with early stages ADPKD. METHODS We recruited 26 ADPKD patients (Group A) with modestly impaired renal function (eGFR 45-70 ml/min/1.73 m(2)), 26 age- and sex-matched ADPKD patients (Group B) with relatively preserved renal function (eGFR)>70 ml/min/1.73 m(2)), and 26 age- and sex-matched controls (Group C). Determination of circulating levels of ADMA, 15-F2t-Isoprostane, oxidized-LDL and routine biochemistry was performed. RESULTS Group A and B had significantly higher ADMA levels as compared to controls (1.68 ± 0.7 vs 0.51 ± 0.2 μmol/l, P<0.001 and 1.26 ± 0.7 vs 0.51 ± 0.2 μmol/l, P<0.001, respectively). 15-F2t-IsoP and oxidized-LDL levels were also significantly higher in Group B relative to controls (788.8 ± 185.0 vs 383.1 ± 86.0 pgr/ml, P<0.001 and 11.4 ± 6.6 vs 6.4 ± 2.6 EU/ml, P<0.05 respectively) and were further elevated in Group A. In correlation analysis, ADMA levels exhibited strong associations with levels of 15-F2t-Isoprostane (r=0.811, P<0.001) and oxidized-LDL (r=0.788, P<0.001), whereas an inverse correlation was evident between ADMA and eGFR (r=-0.460, P<0.001). CONCLUSION This study shows elevation in circulating levels of ADMA along with aggravation of oxidative stress from the early stages of ADPKD. © 2014 S. Karger AG, Basel.
Collapse
Affiliation(s)
- Vassilios Raptis
- Section of Nephrology and Hypertension, 1st Department of Medicine, AHEPA University Hospital, Thessaloniki, Greece
| | | | | | | | | | | | | | | |
Collapse
|
249
|
Kocyigit I, Eroglu E, Orscelik O, Unal A, Gungor O, Ozturk F, Karakukcu C, Imamoglu H, Sipahioglu MH, Tokgoz B, Oymak O. Pentraxin 3 as a novel bio-marker of inflammation and endothelial dysfunction in autosomal dominant polycystic kidney disease. J Nephrol 2014; 27:181-6. [DOI: 10.1007/s40620-014-0045-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 10/10/2013] [Indexed: 02/01/2023]
|
250
|
Yuajit C, Muanprasat C, Gallagher AR, Fedeles SV, Kittayaruksakul S, Homvisasevongsa S, Somlo S, Chatsudthipong V. Steviol retards renal cyst growth through reduction of CFTR expression and inhibition of epithelial cell proliferation in a mouse model of polycystic kidney disease. Biochem Pharmacol 2014; 88:412-21. [PMID: 24518257 DOI: 10.1016/j.bcp.2014.01.038] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Revised: 01/30/2014] [Accepted: 01/31/2014] [Indexed: 01/11/2023]
Abstract
Cyst enlargement in autosomal dominant polycystic kidney disease (ADPKD) is associated with cAMP-activated proliferation of cyst-lining epithelial cells and transepithelial fluid secretion into the cyst lumen via cystic fibrosis transmembrane conductance regulator (CFTR) chloride channel leading to renal failure for which no effective treatment is currently available. We previously reported that steviol retards Madin-Darby canine kidney (MDCK) cyst enlargement by inhibiting CFTR channel activity and promoting proteasomal-mediated CFTR degradation. It is imperative to examine the effect of steviol in animal models of ADPKD. Therefore, we examined the effect of steviol on renal cyst growth in an orthologous mouse model of human ADPKD (Pkd1(flox/flox):Pkhd1-Cre). The results showed that daily treatment with both 200mg/kg BW of steviol and 1000mg/kg BW of stevioside for 14 days markedly decreased kidney weight and cystic index in these mice. However, only steviol markedly reduced blood urea nitrogen and creatinine values. Steviol also reduced cell proliferation but had no effect on cell apoptosis. In addition, steviol suppressed CFTR and mTOR/S6K expression in renal cyst-lining epithelial cells. Interestingly, steviol was found to stimulate AMP-activated protein kinase (AMPK). Our findings indicate that steviol slows cyst progression in ADPKD mouse model, in part, through the activation of AMPK which subsequently inhibits CFTR chloride channel expression and inhibits renal epithelial cell proliferation via mTOR/S6K pathway. Most importantly, steviol could markedly improve kidney function in a mouse model of ADPKD. Steviol thus has potential application for further development as a therapeutic compound for the treatment of polycystic kidney disease.
Collapse
Affiliation(s)
- Chaowalit Yuajit
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Department of Internal Medicine, Genetic, Yale University School of Medicine, New Haven, CT, USA
| | - Chatchai Muanprasat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anna-Rachel Gallagher
- Department of Internal Medicine, Genetic, Yale University School of Medicine, New Haven, CT, USA
| | - Sorin V Fedeles
- Department of Internal Medicine, Genetic, Yale University School of Medicine, New Haven, CT, USA
| | | | - Sureeporn Homvisasevongsa
- Division of Physical Science, Faculty of Science and Technology, Huachiew Chalermprakiet University, Samutprakarn, Thailand
| | - Stefan Somlo
- Department of Internal Medicine, Genetic, Yale University School of Medicine, New Haven, CT, USA.
| | - Varanuj Chatsudthipong
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand; Research Center of Transport Protein for Medical Innovation, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|