1
|
Stanajic-Petrovic G, Keck M, Barbe P, Urman A, Correia E, Isnard P, Duong Van Huyen JP, Chmeis K, Diarra SS, Palea S, Theodoro F, Nguyen AL, Castelli F, Pruvost A, Zhao W, Mendre C, Mouillac B, Bienaimé F, Robin P, Kessler P, Llorens-Cortes C, Servent D, Nozach H, Maillère B, Guo D, Truillet C, Gilles N. A Snake Toxin Derivative for Treatment of Hyponatremia and Polycystic Kidney Diseases. J Am Soc Nephrol 2025; 36:181-192. [PMID: 39431458 PMCID: PMC11801765 DOI: 10.1681/asn.0000000505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 10/02/2024] [Indexed: 10/22/2024] Open
Abstract
Key Points MQ232, a disulfide-bond reticulated peptide derived from a natural snake toxin, was optimized as a new aquaretic drug candidate. MQ232 showed very low acute and chronic toxicity in rat and a biodistribution in mice strongly in favor of the kidney organs. MQ232 induced a sole aquaretic effect and demonstrated high in vivo activities on hyponatremia and polycystic kidney disease models. Background Vaptans were developed at the end of the previous century as vasopressin type 2 receptor antagonists. Tolvaptan is the most prescribed vaptan for hyponatremia and autosomal dominant polycystic kidney disease (ADPKD). However, its use is not as widespread as it should be due to price issues, a narrow therapeutic window, and some side effects. With the aim of discovering new efficient and safer vasopressin type 2 receptor antagonists, we screened animal venoms and identified several peptide toxins. Among them, mambaquaretin 1 (MQ1) displayed unique biological properties in that regard that it was the starting point for the development of a potential drug candidate. Methods Human T-cell assays and bioinformatics were used to mitigate MQ1 immunogenicity risk. MQ232 biodistribution in mice was performed by positron emission tomography. Pharmacodynamics, pharmacokinetics, and acute and chronic toxicity tests were performed on control rats. A rat experimental model of desmopressin-induced hyponatremia, ex vivo mice model of kidney cysts, and mice orthologous model of ADPKD were used to validate MQ232 efficacy in these pathologies. Results Three mutations were introduced in MQ1 to mitigate its immunogenicity risk. A fourth gain-of-function mutation was added to generate MQ232. MQ232's safety was demonstrated by a first toxic dose as high as 3000 nmol/kg and a strong kidney organ selectivity by positron emission tomography imaging, while showing almost no interaction with the liver. MQ232's efficacy was first demonstrated with an effective dose of 3 nmol/kg in a hyponatremic model and then in polycystic kidney models, on which MQ232 significantly reduced cyst growth. Conclusions We demonstrated, using diverse translational techniques and minimizing animal use, MQ232's safety and efficacy in several rodent models of hyponatremia and ADPKD.
Collapse
Affiliation(s)
- Goran Stanajic-Petrovic
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Mathilde Keck
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Peggy Barbe
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Apolline Urman
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Evelyne Correia
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Pierre Isnard
- Anatomie et Cytologie Pathologiques, CHU Necker-Enfants Malades, Paris, France
| | | | - Khawla Chmeis
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | | | - Stefano Palea
- Humana Biosciences, Prologue Biotech, Labège, France
| | - Frederic Theodoro
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Anvi-Laëtitia Nguyen
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Florence Castelli
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Alain Pruvost
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Wenchao Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | | | - Bernard Mouillac
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Frank Bienaimé
- Service d'Explorations Fonctionnelles, Département Croissance et Signalisation, Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Hôpital Necker Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Robin
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Pascal Kessler
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Catherine Llorens-Cortes
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Denis Servent
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Hervé Nozach
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Bernard Maillère
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| | - Dong Guo
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Charles Truillet
- Université Paris-Saclay, CEA, INSERM, CNRS, BioMaps, Orsay, France
| | - Nicolas Gilles
- CEA, Département Médicaments et Technologies pour la Santé (DMTS), Université Paris Saclay, SIMoS, Gif-sur-Yvette, France
| |
Collapse
|
2
|
Parsons AM, Byrne S, Kooistra J, Dewey J, Zebolsky AL, Alvarado G, Bouma GJ, Vanden Heuvel GB, Larson ED. G-quadruplex stabilization provokes DNA breaks in human PKD1, revealing a second hit mechanism for ADPKD. Nat Commun 2025; 16:121. [PMID: 39747084 PMCID: PMC11696556 DOI: 10.1038/s41467-024-55684-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
The "secondhit" pathway is responsible for biallelic inactivation of many tumor suppressors, where a pathogenic germline allele is joined by somatic mutation of the remaining functional allele. The mechanisms are unresolved, but the human PKD1 tumor suppressor is a good experimental model for identifying the molecular determinants. Inactivation of PKD1 results in autosomal dominant polycystic kidney disease, a very common disorder characterized by the accumulation of fluid-filled cysts and end-stage renal disease. Since human PKD1 follows second hit and mouse Pkd1 heterozygotes do not, we reasoned that there is likely a molecular difference that explains the elevated mutagenesis of the human gene. Here we demonstrate that guanine quadruplex DNA structures are abundant throughout human, but not mouse, PKD1 where they activate the DNA damage response. Our results suggest that guanine quadruplex DNAs provoke DNA breaks in PKD1, providing a potential mechanism for cystogenesis in autosomal dominant polycystic kidney disease specifically and for the inactivation of guanine quadruplex-rich tumor suppressors generally.
Collapse
Affiliation(s)
- Agata M Parsons
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Seth Byrne
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Jesse Kooistra
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - John Dewey
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Aaron L Zebolsky
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Gloria Alvarado
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Gerrit J Bouma
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Gregory B Vanden Heuvel
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA
| | - Erik D Larson
- Department of Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, MI, USA.
| |
Collapse
|
3
|
Zhou L, Wei X, Wang B, Xu Q, Li W. Low-dose tolvaptan to control disease progression in Chinese patients with autosomal dominant polycystic kidney disease: a retrospective cohort study. Transl Androl Urol 2024; 13:2307-2321. [PMID: 39507867 PMCID: PMC11535741 DOI: 10.21037/tau-24-448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Background Tolvaptan has been shown to be effective in the treatment of autosomal dominant polycystic kidney disease (ADPKD). However, there is limited evidence regarding optimal dosing and its application within the Chinese population. In this study, we aimed to determine whether a lower tolvaptan dose could effectively control ADPKD in Chinese patients. Methods This retrospective, single-center cohort study was conducted in a real-world setting and included all patients newly diagnosed with rapidly progressive ADPKD who initiated tolvaptan treatment and maintained it for at least 12 months. Data were collected at baseline and at 1, 2, 4, 8, and 12 months after treatment initiation. Patients began with morning/evening tolvaptan doses of 7.5 mg/7.5 mg, and the dose was subsequently adjusted based on effectiveness and tolerability. The patients were categorized by baseline estimated glomerular filtration rate (eGFR) and final daily tolvaptan dose. Changes in eGFR and other key physiological indicators after treatment were compared within each group. Results The study included 43 patients with ADPKD, of whom 20 were female, with a median age of 34.3 years (range, 16-85 years). At 12 months, eGFR improved by 5.48 mL/min/1.73 m2 [95% confidence interval (CI): 2.68-8.29] (P<0.001) compared to baseline. Significant improvements were observed in patients with baseline eGFR levels of 30-59, 60-89, and ≥90 mL/min/1.73 m2 (P=0.007, 0.045, and 0.02, respectively), as well as in medium and high dose groups (P=0.002 and 0.02, respectively). At 12 months, the annual height-adjusted total kidney volume (HtTKV) growth slope decreased by -0.17 %/year (95% CI: -0.33 to -0.01) (P=0.04). Significant decreases were observed in patients with an eGFR of 30-59 mL/min/1.73 m2 (P=0.008) and in the medium dose group (P=0.03). Thirst was reported in 22 (51.2%) patients, all of whom experienced mild symptoms. No liver-associated adverse events were noted. Conclusions Tolvaptan is well tolerated at low initial doses in Chinese patients with ADPKD. Significant improvements in eGFR and reduced HtTKV growth were observed in the overall population and across various baseline eGFR and final dose groups.
Collapse
Affiliation(s)
- Li Zhou
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Xiansen Wei
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Boya Wang
- Department of Ultrasound, China-Japan Friendship Hospital, Beijing, China
| | - Qianqian Xu
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| | - Wenge Li
- Department of Nephrology, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
4
|
Lv J, Lan B, Fu L, He C, Zhou W, Wang X, Zhou C, Mao Z, Chen Y, Mei C, Xue C. EZH2 inhibition or genetic ablation suppresses cyst growth in autosomal dominant polycystic kidney disease. J Transl Med 2024; 22:979. [PMID: 39472935 PMCID: PMC11520870 DOI: 10.1186/s12967-024-05785-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 10/18/2024] [Indexed: 11/02/2024] Open
Abstract
BACKGROUND Autosomal Dominant Polycystic Kidney Disease (ADPKD) is a prevalent genetic disorder characterized by the formation of renal cysts leading to kidney failure. Despite known genetic underpinnings, the variability in disease progression suggests additional regulatory layers, including epigenetic modifications. METHODS We utilized various ADPKD models, including Pkd1 and Ezh2 conditional knockout (Pkd1delta/delta:Ezh2delta/delta) mice, to explore the role of Enhancer of Zeste Homolog 2 (EZH2) in cystogenesis. Pharmacological inhibition of EZH2 was performed using GSK126 or EPZ-6438 across multiple models. RESULTS EZH2 expression was significantly upregulated in Pkd1-/- cells, Pkd1delta/delta mice, and human ADPKD kidneys. EZH2 inhibition attenuates cyst development in MDCK cells and a mouse embryonic kidney cyst model. Both Ezh2 conditional knockout and GSK126 treatment suppressed renal cyst growth and protected renal function in Pkd1delta/delta mice. Mechanistically, cAMP/PKA/CREB pathway increased EZH2 expression. EZH2 mediated cystogenesis by enhancing methylation and activation of STAT3, promoting cell cycle through p21 suppression, and stimulating non-phosphorylated β-catenin in Wnt signaling pathway. Additionally, EZH2 enhanced ferroptosis by inhibiting SLC7A11 and GPX4 in ADPKD. CONCLUSION Our findings elucidate the pivotal role of EZH2 in promoting renal cyst growth through epigenetic mechanisms and suggest that EZH2 inhibition or ablation may serve as a novel therapeutic approach for managing ADPKD.
Collapse
Affiliation(s)
- Jiayi Lv
- Kidney Institute, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), 415 Fengyang Road, Shanghai, 200003, China
| | - Bingxue Lan
- Center for Clinical Laboratories, Affiliated Hospital of Guizhou Medical University, Guiyang City, Guizhou Province, China
- Department of Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Lili Fu
- Kidney Institute, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), 415 Fengyang Road, Shanghai, 200003, China
| | - Chaoran He
- Department of Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China
| | - Wei Zhou
- Department of Nephrology, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xi Wang
- Institute of Infectious Diseases, Beijing Key Laboratory of Emerging Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
- Beijing Institute of Infectious Diseases, Beijing, 100015, China
| | - Chenchen Zhou
- Outpatient Department, Yangpu Third Military Retreat, Shanghai, China
| | - Zhiguo Mao
- Kidney Institute, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), 415 Fengyang Road, Shanghai, 200003, China.
| | - Yupeng Chen
- Department of Biochemistry and Molecular Biology, 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Tianjin Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, Tianjin Medical University, 22 Qixiangtai Road, Tianjin, 300070, China.
| | - Changlin Mei
- Kidney Institute, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), 415 Fengyang Road, Shanghai, 200003, China.
| | - Cheng Xue
- Kidney Institute, Division of Nephrology, Shanghai Changzheng Hospital, Second Military Medical University (Naval Medical University), 415 Fengyang Road, Shanghai, 200003, China.
| |
Collapse
|
5
|
Hong S, Song M, Miyoshi T, Morizane R, Bonventre JV, Lee LP. Dynamic Kidney Organoid Microphysiological Analysis Platform. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.27.620552. [PMID: 39554191 PMCID: PMC11565751 DOI: 10.1101/2024.10.27.620552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Kidney organoids, replicating human development, pathology, and drug responses, are a promising model for advancing bioscience and pharmaceutical innovation. However, reproducibility, accuracy, and quantification challenges hinder their broader utility for advanced biological and pharmaceutical applications. Herein, we present a dynamic kidney organoid microphysiological analysis platform (MAP), designed to enhance organoid modeling and assays within physiologically relevant environments, thereby expanding their utility in advancing kidney physiology and pathology research. First, precise control of the dynamic microenvironment in MAP enhances the ability to fine-tune nephrogenic intricacies, facilitating high-throughput and reproducible human kidney organoid development. Also, MAP's miniaturization of kidney organoids significantly advances pharmaceutical research by allowing for detailed analysis of entire nephron segments, which is crucial for assessing the nephrotoxicity and safety of drugs. Furthermore, the MAP's application in disease modeling faithfully recapitulates pathological development and functions as a valuable testbed for therapeutic exploration in polycystic kidney diseases. We envision the kidney organoid MAP enhancing pharmaceutical research, standardizing processes, and improving analytics, thereby elevating the quality and utility of organoids in biology, pharmacology, precision medicine, and education.
Collapse
Affiliation(s)
- SoonGweon Hong
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Minsun Song
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomoya Miyoshi
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryuji Morizane
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Massachusetts General Hospital, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge and Boston, Boston, MA, USA
| | - Joseph V. Bonventre
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- John A. Paulson School of Engineering and Applied Sciences and Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge and Boston, Boston, MA, USA
| | - Luke P. Lee
- Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Department of Bioengineering, Department of Electrical Engineering and Computer Science, University of California at Berkeley, Berkeley, CA, USA
- Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Korea
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, Korea
| |
Collapse
|
6
|
Devi C, Ranjan P, Raj S, Das P. Computational exploration of protein structure dynamics and RNA structural consequences of PKD1 missense variants: implications in ADPKD pathogenesis. 3 Biotech 2024; 14:211. [PMID: 39188533 PMCID: PMC11344749 DOI: 10.1007/s13205-024-04057-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
We analyzed the impact of nine previously identified missense PKD1 variants from our studies, including c.6928G > A p.G2310R, c.8809G > A p.E2937K, c.2899 T > C p.W967R, c.6284A > G p.D2095G, c.6644G > A p.R2215Q, c.7810G > A p.D2604N, c.11249G > C p.R3750P, c.1001C > T p.T334M, and c.3101A > G p.N1034S on RNA structures and PC1 protein structure dynamics utilizing computational tools. RNA structure analysis was done using short RNA snippets of 41 nucleotides with the variant position at the 21st nucleotide, ensuring 20 bases on both sides. The secondary structures of these RNA snippets were predicted using RNAstructure. Structural changes of the mutants compared to the wild type were analyzed using the MutaRNA webserver. Molecular dynamics (MD) simulation of PC1 wild-type and mutant protein regions were performed using GROMACS 2018 (GROMOS96 54a7 force field). Findings revealed that five variants including c.8809G > A (p.E2937K), c.11249G > C (p.R3750P), c.3101A > G (p.N1034S), c.6928G > A (p.G2310R), c.6644G > A (p.R2215Q) exhibited major alterations in RNA structures and thereby their interactions with other proteins or RNAs affecting protein structure dynamics. While certain variants have minimal impact on RNA conformations, their observed alterations in MD simulations indicate impact on protein structure dynamics highlighting the importance of evaluating the functional consequences of genetic variants by considering both RNA and protein levels. The study also emphasizes that each missense variant exerts a unique impact on RNA stability, and protein structure dynamics, potentially contributing to the heterogeneous clinical manifestations and progression observed in Autosomal Dominant Polycystic Kidney Disease (ADPKD) patients offering a novel perspective in this direction. Thus, the utility of studying the structure dynamics through computational tools can help in prioritizing the variants for their functional implications, understanding the molecular mechanisms underlying variability in ADPKD presentation and developing targeted therapeutic interventions. Supplementary Information The online version contains supplementary material available at 10.1007/s13205-024-04057-9.
Collapse
Affiliation(s)
- Chandra Devi
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005 India
| | - Prashant Ranjan
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005 India
| | - Sonam Raj
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| | - Parimal Das
- Centre for Genetic Disorders, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh 221005 India
| |
Collapse
|
7
|
Puri B, Kulkarni YA, Gaikwad AB. Advances in CRISPR-Cas systems for kidney diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 210:149-162. [PMID: 39824579 DOI: 10.1016/bs.pmbts.2024.07.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2025]
Abstract
Recent advances in CRISPR-Cas systems have revolutionised the study and treatment of kidney diseases, including acute kidney injury (AKI), chronic kidney disease (CKD), diabetic kidney disease (DKD), lupus nephritis (LN), and polycystic kidney disease (PKD). CRISPR-Cas technology offers precise and versatile tools for genetic modification in monogenic kidney disorders such as PKD and Alport syndrome. Recent advances in CRISPR technology have also shown promise in addressing other kidney diseases like AKI, CKD, and DKD. CRISPR-Cas holds promise to edit genetic mutations underlying these conditions, potentially leading to more effective and long-lasting treatments. Furthermore, the adaptability of CRISPR-Cas systems allows for developing tailored therapeutic strategies that specifically target the genetic and molecular mechanisms contributing to different kidney diseases. Beyond DNA modifications, CRISPR-Cas technologies also enable editing noncoding RNA, such as lncRNAs and miRNAs, in kidney diseases. Despite these advancements, significant challenges persist, including delivery efficiency to specific kidney cells and potential off-target effects. However, the rapid progress in CRISPR-Cas technology suggests a transformative impact on the future management of kidney diseases, offering the potential for enhanced patient outcomes through personalised and precise therapeutic approaches. This chapter highlights the recent advancement of CRISPR-Cas systems and their potential applications in various kidney diseases.
Collapse
Affiliation(s)
- Bhupendra Puri
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta road, Vile Parle (W), Mumbai, India
| | - Anil Bhanudas Gaikwad
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India.
| |
Collapse
|
8
|
Ndongo M, Nehemie LM, Coundoul B, Diouara AAM, Seck SM. Prevalence and outcomes of polycystic kidney disease in African populations: A systematic review. World J Nephrol 2024; 13:90402. [PMID: 38596265 PMCID: PMC11000041 DOI: 10.5527/wjn.v13.i1.90402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/03/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024] Open
Abstract
BACKGROUND Polycystic kidney disease (PKD) is the most common genetic cause of kidney disease. It is a progressive and irreversible condition that can lead to end-stage renal disease and many other visceral complications. Current comprehensive data on PKD patterns in Africa is lacking. AIM To describe the prevalence and outcomes of PKD in the African population. METHODS A literature search of PubMed, African journal online, and Google Scholar databases between 2000 and 2023 was performed. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses were followed to design the study. Clinical presentations and outcomes of patients were extracted from the included studies. RESULTS Out of 106 articles, we included 13 studies from 7 African countries. Ten of them were retrospective descriptive studies concerning 943 PKD patients with a mean age of 47.9 years. The accurate prevalence and incidence of PKD were not known but it represented the third causal nephropathy among dialysis patients. In majority of patients, the diagnosis of the disease was often delayed. Kidney function impairment, abdominal mass, and hypertension were the leading symptoms at presentation with a pooled prevalence of 72.1% (69.1-75.1), 65.8% (62.2-69.4), and 57.4% (54.2-60.6) respectively. Hematuria and infections were the most frequent complications. Genotyping was performed in few studies that revealed a high proportion of new mutations mainly in the PKD1 gene. CONCLUSION The prevalence of PKD in African populations is not clearly defined. Clinical symptoms were almost present with most patients who had kidney function impairment and abdominal mass at the diagnostic. Larger studies including genetic testing are needed to determine the burden of PKD in African populations.
Collapse
Affiliation(s)
- Modou Ndongo
- Department of Nephrology and Dialysis, Regional Hospital of Kedougou, Kedougou 26005, Senegal
| | - Lot Motoula Nehemie
- Department of Nephrology and Dialysis, Military Hospital of Ouakam, Dakar 28216, Senegal
| | - Baratou Coundoul
- Department of Nephrology and Dialysis, Military Hospital of Ouakam, Dakar 28216, Senegal
| | - Abou Abdallah Malick Diouara
- Department of Chemical Engineering and Applied Biology, Polytechnic high School of Cheikh Anta Diop University, Dakar 5085, Senegal
| | - Sidy Mohamed Seck
- Department of Nephrology and Dialysis, Military Hospital of Ouakam, Dakar 28216, Senegal
- Department of Nephrology, Faculty of Health Sciences, University Gaston Berger, Saint-Louis 234, Senegal
| |
Collapse
|
9
|
Liu Y, Zhu P, Tian J. Case report: Rare genetic liver disease - a case of congenital hepatic fibrosis in adults with autosomal dominant polycystic kidney disease. Front Med (Lausanne) 2024; 11:1344151. [PMID: 38384417 PMCID: PMC10879390 DOI: 10.3389/fmed.2024.1344151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
Congenital hepatic fibrosis (CHF) is considered to be a rare autosomal recessive hereditary fibrocystic liver disease, mainly found in children. However, cases of adult CHF with autosomal dominant polycystic kidney disease (ADPKD) caused by PKD1 gene mutation are extremely rare. We report a 31-year-old female patient admitted for esophageal and gastric variceal bleeding. Physical examination revealed significant splenomegaly, biochemical tests showed a slight increase in liver enzymes, and a decrease in platelet count. Imaging examinations showed significant dilatation of the common bile duct and intrahepatic bile ducts, as well as multiple renal cysts. Liver biopsy revealed enlarged portal areas, bridging fibrosis, and numerous variably shaped small bile ducts. Genetic testing identified two unique mutations in the PKD1 gene, identified as biallelic mutations compound heterozygous mutations composed of a mutation inherited from the father (c.8296 T > C) and one from the mother (c.9653G > C). Based on multiple test results, the patient was diagnosed with the portal hypertension type CHF associated with ADPKD. During her initial hospital stay, the patient underwent endoscopic treatment for gastrointestinal bleeding. To date, the patient has recovered well. Moreover, a significant reduction in varices was observed in a gastroscopy examination 18 months later.
Collapse
Affiliation(s)
- Ying Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Ping Zhu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Jiajun Tian
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
10
|
Kofotolios I, Bonios MJ, Adamopoulos M, Mourouzis I, Filippatos G, Boletis JN, Marinaki S, Mavroidis M. The Han:SPRD Rat: A Preclinical Model of Polycystic Kidney Disease. Biomedicines 2024; 12:362. [PMID: 38397964 PMCID: PMC10887417 DOI: 10.3390/biomedicines12020362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) stands as the most prevalent hereditary renal disorder in humans, ultimately culminating in end-stage kidney disease. Animal models carrying mutations associated with polycystic kidney disease have played an important role in the advancement of ADPKD research. The Han:SPRD rat model, carrying an R823W mutation in the Anks6 gene, is characterized by cyst formation and kidney enlargement. The mutated protein, named Samcystin, is localized in cilia of tubular epithelial cells and seems to be involved in cystogenesis. The homozygous Anks6 mutation leads to end-stage renal disease and death, making it a critical factor in kidney development and function. This review explores the utility of the Han:SPRD rat model, highlighting its phenotypic similarity to human ADPKD. Specifically, we discuss its role in preclinical trials and its importance for investigating the pathogenesis of the disease and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Ioannis Kofotolios
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| | - Michael J. Bonios
- Heart Failure and Transplant Unit, Onassis Cardiac Surgery Center, 17674 Athens, Greece;
| | - Markos Adamopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Gerasimos Filippatos
- Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - John N. Boletis
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Smaragdi Marinaki
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| |
Collapse
|
11
|
Parsons AM, Su K, Daniels M, Bouma GJ, Vanden Heuvel GB, Larson ED. Human PKD1 sequences form R-loop structures in vitro. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001058. [PMID: 38371318 PMCID: PMC10873753 DOI: 10.17912/micropub.biology.001058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/08/2024] [Accepted: 01/30/2024] [Indexed: 02/20/2024]
Abstract
Autosomal dominant polycystic kidney disease results from the loss of the PKD1 gene product, polycystin 1. Regulatory mechanisms are unresolved, but an apparent G/C sequence bias in the gene is consistent with co-transcriptional R-loop formation. R-loops regulate gene expression and stability, and they form when newly synthesized RNA extensively pairs with the template DNA to displace the non-template strand. In this study, we tested two human PKD1 sequences for co-transcriptional R-loop formation in vitro. We observed RNase H-sensitive R-loop formation in intron 1 and 22 sequences, but only in one transcriptional orientation. Therefore, R-loops may participate in PKD1 expression or stability.
Collapse
Affiliation(s)
- Agata M Parsons
- Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, United States
| | - Kemin Su
- Investigative Medicine, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, United States
| | - Maya Daniels
- Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, United States
| | - Gerrit J Bouma
- Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, United States
| | - Gregory B Vanden Heuvel
- Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, United States
| | - Erik D Larson
- Biomedical Sciences, Western Michigan University Homer Stryker MD School of Medicine, Kalamazoo, Michigan, United States
| |
Collapse
|
12
|
黄 丹, 刘 雅, 李 丹, 张 静, 杨 翌, 孙 良. [C/EBPβ mediates expressions of downstream inflammatory factors of the tumor necrosis factor- α signaling pathway in renal tubular epithelial cells with NPHP1 knockdown]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2024; 44:156-165. [PMID: 38293987 PMCID: PMC10878891 DOI: 10.12122/j.issn.1673-4254.2024.01.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Indexed: 02/01/2024]
Abstract
OBJECTIVE To explore the activation of tumor necrosis factor-α (TNF-α) signaling pathway and the expressions of the associated inflammatory factors in NPHP1-defective renal tubular epithelial cells. METHODS A human proximal renal tubular cell (HK2) model of lentivirus-mediated NPHP1 knockdown (NPHP1KD) was constructed, and the expressions of TNF-α, p38, and C/EBPβ and the inflammatory factors CXCL5, CCL20, IL-1β, IL-6 and MCP-1 were detected using RT-qPCR, Western blotting or enzyme-linked immunosorbent assay. A small interfering RNA (siRNA) was transfected in wild-type and NPHP1KDHK2 cells, and the changes in the expressions of TNF-α, p38, and C/EBPβ and the inflammatory factors were examined. RESULTS NPHP1KDHK2 cells showed significantly increased mRNA expressions of TNF-α, C/EBPβ, CXCL5, IL-1β, and IL-6 (P < 0.05), protein expressions of phospho-p38 and C/EBPβ (P < 0.05), and IL-6 level in the culture supernatant (P < 0.05), and these changes were significantly blocked by transfection of cells with siRNA-C/EBPβ (P < 0.05). CONCLUSION TNF-α signaling pathway is activated and its associated inflammatory factors are upregulated in NPHP1KDHK2 cells, and C/EBPβ may serve as a key transcription factor to mediate these changes.
Collapse
Affiliation(s)
- 丹梅 黄
- />南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 雅清 刘
- />南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 丹彤 李
- />南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 静兰 张
- />南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 翌晨 杨
- />南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 良忠 孙
- />南方医科大学南方医院儿科,广东 广州 510515Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
13
|
Ateshian GA, Spack KA, Hone JC, Azeloglu EU, Gusella GL. Computational study of biomechanical drivers of renal cystogenesis. Biomech Model Mechanobiol 2023; 22:1113-1127. [PMID: 37024601 PMCID: PMC10524738 DOI: 10.1007/s10237-023-01704-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 02/12/2023] [Indexed: 04/08/2023]
Abstract
Renal cystogenesis is the pathological hallmark of autosomal dominant polycystic kidney disease, caused by PKD1 and PKD2 mutations. The formation of renal cysts is a common manifestation in ciliopathies, a group of syndromic disorders caused by mutation of proteins involved in the assembly and function of the primary cilium. Cystogenesis is caused by the derailment of the renal tubular architecture and tissue deformation that eventually leads to the impairment of kidney function. However, the biomechanical imbalance of cytoskeletal forces that are altered in cells with Pkd1 mutations has never been investigated, and its nature and extent remain unknown. In this computational study, we explored the feasibility of various biomechanical drivers of renal cystogenesis by examining several hypothetical mechanisms that may promote morphogenetic markers of cystogenesis. Our objective was to provide physics-based guidance for our formulation of hypotheses and our design of experimental studies investigating the role of biomechanical disequilibrium in cystogenesis. We employed the finite element method to explore the role of (1) wild-type versus mutant cell elastic modulus; (2) contractile stress magnitude in mutant cells; (3) localization and orientation of contractile stress in mutant cells; and (4) sequence of cell contraction and cell proliferation. Our objective was to identify the factors that produce the characteristic tubular cystic growth. Results showed that cystogenesis occurred only when mutant cells contracted along the apical-basal axis, followed or accompanied by cell proliferation, as long as mutant cells had comparable or lower elastic modulus than wild-type cells, with their contractile stresses being significantly greater than their modulus. Results of these simulations allow us to focus future in vitro and in vivo experimental studies on these factors, helping us formulate physics-based hypotheses for renal tubule cystogenesis.
Collapse
Affiliation(s)
- Gerard A Ateshian
- Department of Mechanical Engineering, Columbia University, New York, NY, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, USA.
| | - Katherine A Spack
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - James C Hone
- Department of Mechanical Engineering, Columbia University, New York, NY, USA
| | - Evren U Azeloglu
- Department of Medicine, Division of Nephrology, Mount Sinai School of Medicine, New York, NY, USA
- Department of Pharmacological Sciences, Mount Sinai School of Medicine, New York, NY, USA
| | - G Luca Gusella
- Department of Medicine, Division of Nephrology, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
14
|
Dagorn PG, Buchholz B, Kraus A, Batchuluun B, Bange H, Blockken L, Steinberg GR, Moller DE, Hallakou-Bozec S. A novel direct adenosine monophosphate kinase activator ameliorates disease progression in preclinical models of Autosomal Dominant Polycystic Kidney Disease. Kidney Int 2023; 103:917-929. [PMID: 36804411 DOI: 10.1016/j.kint.2023.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 01/13/2023] [Accepted: 01/27/2023] [Indexed: 02/21/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) mainly results from mutations in the PKD1 gene, which encodes polycystin 1. It is the most common inherited kidney disease and is characterized by a progressive bilateral increase in cyst number and size, often leading to kidney failure. The cellular energy sensor and regulator adenosine monophosphate stimulated protein kinase (AMPK) has been implicated as a promising new therapeutic target. To address this hypothesis, we determined the effects of a potent and selective clinical stage direct allosteric AMPK activator, PXL770, in canine and patient-derived 3D cyst models and an orthologous mouse model of ADPKD. PXL770 induced AMPK activation and dose-dependently reduced cyst growth in principal-like Madin-Darby Canine Kidney cells stimulated with forskolin and kidney epithelial cells derived from patients with ADPKD stimulated with desmopressin. In an inducible, kidney epithelium-specific Pkd1 knockout mouse model, PXL770 produced kidney AMPK pathway engagement, prevented the onset of kidney failure (reducing blood urea by 47%), decreased cystic index by 26% and lowered the kidney weight to body weight ratio by 35% compared to untreated control Pkd1 knockout mice. These effects were accompanied by a reduction of markers of cell proliferation (-48%), macrophage infiltration (-53%) and tissue fibrosis (-37%). Thus, our results show the potential of direct allosteric AMPK activation in the treatment of ADPKD and support the further development of PXL770 for this indication.
Collapse
Affiliation(s)
| | - Bjoern Buchholz
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andre Kraus
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Battsetseg Batchuluun
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Hester Bange
- Crown Bioscience Netherlands B.V., The Netherlands
| | | | - Gregory R Steinberg
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine and Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
15
|
Li Z, Zimmerman KA, Cherakara S, Chumley PH, Collawn JF, Wang J, Haycraft CJ, Song CJ, Chacana T, Andersen RS, Croyle MJ, Aloria EJ, Hombal RP, Thomas IN, Chweih H, Simanyi KL, George JF, Parant JM, Mrug M, Yoder BK. A kidney resident macrophage subset is a candidate biomarker for renal cystic disease in preclinical models. Dis Model Mech 2023; 16:dmm049810. [PMID: 36457161 PMCID: PMC9884121 DOI: 10.1242/dmm.049810] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 11/21/2022] [Indexed: 12/04/2022] Open
Abstract
Although renal macrophages have been shown to contribute to cyst development in polycystic kidney disease (PKD) animal models, it remains unclear whether there is a specific macrophage subpopulation involved. Here, we analyzed changes in macrophage populations during renal maturation in association with cystogenesis rates in conditional Pkd2 mutant mice. We observed that CD206+ resident macrophages were minimal in a normal adult kidney but accumulated in cystic areas in adult-induced Pkd2 mutants. Using Cx3cr1 null mice, we reduced macrophage number, including CD206+ macrophages, and showed that this significantly reduced cyst severity in adult-induced Pkd2 mutant kidneys. We also found that the number of CD206+ resident macrophage-like cells increased in kidneys and in the urine from autosomal-dominant PKD (ADPKD) patients relative to the rate of renal functional decline. These data indicate a direct correlation between CD206+ resident macrophages and cyst formation, and reveal that the CD206+ resident macrophages in urine could serve as a biomarker for renal cystic disease activity in preclinical models and ADPKD patients. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Zhang Li
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kurt A. Zimmerman
- Division of Nephrology, Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 732104, USA
| | - Sreelakshmi Cherakara
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Phillip H. Chumley
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - James F. Collawn
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jun Wang
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Courtney J. Haycraft
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Cheng J. Song
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Teresa Chacana
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Reagan S. Andersen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Mandy J. Croyle
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ernald J. Aloria
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Raksha P. Hombal
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Isis N. Thomas
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hanan Chweih
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kristin L. Simanyi
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - James F. George
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Michal Mrug
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
- Department of Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, AL 35233, USA
| | - Bradley K. Yoder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
16
|
Zhang Z, Blumenfeld J, Ramnauth A, Barash I, Zhou P, Levine D, Parker T, Rennert H. A common intronic single nucleotide variant modifies PKD1 expression level. Clin Genet 2022; 102:483-493. [PMID: 36029107 PMCID: PMC10947153 DOI: 10.1111/cge.14214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/20/2022] [Accepted: 08/22/2022] [Indexed: 11/26/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), caused by mutations in PKD1 and PKD2 (PKD1/2), has unexplained phenotypic variability likely affected by environmental and other genetic factors. Approximately 10% of individuals with ADPKD phenotype have no causal mutation detected, possibly due to unrecognized risk variants of PKD1/2. This study was designed to identify risk variants of PKD genes through population genetic analyses. We used Wright's F-statistics (Fst) to evaluate common single nucleotide variants (SNVs) potentially favored by positive natural selection in PKD1 from 1000 Genomes Project (1KG) and genotyped 388 subjects from the Rogosin Institute ADPKD Data Repository. The variants with >90th percentile Fst scores underwent further investigation by in silico analysis and molecular genetics analyses. We identified a deep intronic SNV, rs3874648G> A, located in a conserved binding site of the splicing regulator Tra2-β in PKD1 intron 30. Reverse-transcription PCR (RT-PCR) of peripheral blood leukocytes (PBL) from an ADPKD patient homozygous for rs3874648-A identified an atypical PKD1 splice form. Functional analyses demonstrated that rs3874648-A allele increased Tra2-β binding affinity and activated a cryptic acceptor splice-site, causing a frameshift that introduced a premature stop codon in mRNA, thereby decreasing PKD1 full-length transcript level. PKD1 transcript levels were lower in PBL from rs3874648-G/A carriers than in rs3874648-G/G homozygotes in a small cohort of normal individuals and patients with PKD2 inactivating mutations. Our findings indicate that rs3874648G > A is a PKD1 expression modifier attenuating PKD1 expression through Tra2-β, while the derived G allele advantageously maintains PKD1 expression and is predominant in all subpopulations.
Collapse
Affiliation(s)
- Zhengmao Zhang
- Departments of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Jon Blumenfeld
- Department of Medicine, Weill Cornell Medicine, New York, NY
- The Rogosin Institute, New York, NY
| | - Andrew Ramnauth
- Departments of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Irina Barash
- Department of Medicine, Weill Cornell Medicine, New York, NY
- The Rogosin Institute, New York, NY
| | - Pengbo Zhou
- Departments of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| | - Daniel Levine
- Department of Biochemistry, Weill Cornell Medicine, New York, NY
- The Rogosin Institute, New York, NY
| | - Thomas Parker
- Department of Biochemistry, Weill Cornell Medicine, New York, NY
- The Rogosin Institute, New York, NY
| | - Hanna Rennert
- Departments of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY
| |
Collapse
|
17
|
Wang JY, Wang J, Lu XG, Song W, Luo S, Zou DF, Hua LD, Peng Q, Tian Y, Gao LD, Liao WP, He N. Recessive PKD1 Mutations Are Associated With Febrile Seizures and Epilepsy With Antecedent Febrile Seizures and the Genotype-Phenotype Correlation. Front Mol Neurosci 2022; 15:861159. [PMID: 35620448 PMCID: PMC9128595 DOI: 10.3389/fnmol.2022.861159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/05/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveThe PKD1 encodes polycystin-1, a large transmembrane protein that plays important roles in cell proliferation, apoptosis, and cation transport. Previous studies have identified PKD1 mutations in autosomal dominant polycystic kidney disease (ADPKD). However, the expression of PKD1 in the brain is much higher than that in the kidney. This study aimed to explore the association between PKD1 and epilepsy.MethodsTrios-based whole-exome sequencing was performed in a cohort of 314 patients with febrile seizures or epilepsy with antecedent febrile seizures. The damaging effects of variants was predicted by protein modeling and multiple in silico tools. The genotype-phenotype association of PKD1 mutations was systematically reviewed and analyzed.ResultsEight pairs of compound heterozygous missense variants in PKD1 were identified in eight unrelated patients. All patients suffered from febrile seizures or epilepsy with antecedent febrile seizures with favorable prognosis. All of the 16 heterozygous variants presented no or low allele frequencies in the gnomAD database, and presented statistically higher frequency in the case-cohort than that in controls. These missense variants were predicted to be damaging and/or affect hydrogen bonding or free energy stability of amino acids. Five patients showed generalized tonic-clonic seizures (GTCS), who all had one of the paired missense mutations located in the PKD repeat domain, suggesting that mutations in the PKD domains were possibly associated with GTCS. Further analysis demonstrated that monoallelic mutations with haploinsufficiency of PKD1 potentially caused kidney disease, compound heterozygotes with superimposed effects of two missense mutations were associated with epilepsy, whereas the homozygotes with complete loss of PKD1 would be embryonically lethal.ConclusionPKD1 gene was potentially a novel causative gene of epilepsy. The genotype-phenotype relationship of PKD1 mutations suggested a quantitative correlation between genetic impairment and phenotypic variation, which will facilitate the genetic diagnosis and management in patients with PKD1 mutations.
Collapse
Affiliation(s)
- Jing-Yang Wang
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Jie Wang
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Xin-Guo Lu
- Epilepsy Center, Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Wang Song
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Sheng Luo
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Dong-Fang Zou
- Epilepsy Center, Department of Neurology, Shenzhen Children’s Hospital, Shenzhen, China
| | - Li-Dong Hua
- Translational Medicine Center, Guangdong Women and Children Hospital, Guangzhou, China
| | - Qian Peng
- Department of Pediatrics, Dongguan City Maternal and Child Health Hospital, Southern Medical University, Dongguan, China
| | - Yang Tian
- Department of Neurology, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Liang-Di Gao
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Wei-Ping Liao
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
| | - Na He
- Department of Neurology, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Key Laboratory of Neurogenetics and Channelopathies of the Ministry of Education of China, Guangzhou, China
- *Correspondence: Na He,
| |
Collapse
|
18
|
Carotti V, van der Wijst J, Verschuren EHJ, Rutten L, Sommerdijk N, Kaffa C, Sommers V, Rigalli JP, Hoenderop JGJ. Involvement of ceramide biosynthesis in increased extracellular vesicle release in Pkd1 knock out cells. Front Endocrinol (Lausanne) 2022; 13:1005639. [PMID: 36299464 PMCID: PMC9589111 DOI: 10.3389/fendo.2022.1005639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is an inherited disorder characterized by the development of renal cysts, which frequently leads to renal failure. Hypertension and other cardiovascular symptoms contribute to the high morbidity and mortality of the disease. ADPKD is caused by mutations in the PKD1 gene or, less frequently, in the PKD2 gene. The disease onset and progression are highly variable between patients, whereby the underlying mechanisms are not fully elucidated. Recently, a role of extracellular vesicles (EVs) in the progression of ADPKD has been postulated. However, the mechanisms stimulating EV release in ADPKD have not been addressed and the participation of the distal nephron segments is still uninvestigated. Here, we studied the effect of Pkd1 deficiency on EV release in wild type and Pkd1-/- mDCT15 and mIMCD3 cells as models of the distal convoluted tubule (DCT) and inner medullary collecting duct (IMCD), respectively. By using nanoparticle tracking analysis, we observed a significant increase in EV release in Pkd1-/- mDCT15 and mIMCD3 cells, with respect to the wild type cells. The molecular mechanisms leading to the changes in EV release were further investigated in mDCT15 cells through RNA sequencing and qPCR studies. Specifically, we assessed the relevance of purinergic signaling and ceramide biosynthesis enzymes. Pkd1-/- mDCT15 cells showed a clear upregulation of P2rx7 expression compared to wild type cells. Depletion of extracellular ATP by apyrase (ecto-nucleotidase) inhibited EV release only in wild type cells, suggesting an exacerbated signaling of the extracellular ATP/P2X7 pathway in Pkd1-/- cells. In addition, we identified a significant up-regulation of the ceramide biosynthesis enzymes CerS6 and Smpd3 in Pkd1-/- cells. Altogether, our findings suggest the involvement of the DCT in the EV-mediated ADPKD progression and points to the induction of ceramide biosynthesis as an underlying molecular mechanism. Further studies should be performed to investigate whether CerS6 and Smpd3 can be used as biomarkers of ADPKD onset, progression or severity.
Collapse
Affiliation(s)
- Valentina Carotti
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Jenny van der Wijst
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Eric H. J. Verschuren
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Luco Rutten
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Nico Sommerdijk
- Electron Microscopy Center, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Charlotte Kaffa
- Radboud Technology Center for Bioinformatics, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Vera Sommers
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Juan P. Rigalli
- Department of Clinical Pharmacology and Pharmacoepidemiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Joost G. J. Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
- *Correspondence: Joost G. J. Hoenderop,
| |
Collapse
|
19
|
Zhang Z, Bai H, Blumenfeld J, Ramnauth AB, Barash I, Prince M, Tan AY, Michaeel A, Liu G, Chicos I, Rennert L, Giannakopoulos S, Larbi K, Hughes S, Salvatore SP, Robinson BD, Kapur S, Rennert H. Detection of PKD1 and PKD2 Somatic Variants in Autosomal Dominant Polycystic Kidney Cyst Epithelial Cells by Whole-Genome Sequencing. J Am Soc Nephrol 2021; 32:3114-3129. [PMID: 34716216 PMCID: PMC8638386 DOI: 10.1681/asn.2021050690] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 09/03/2021] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is a genetic disorder characterized by the development of multiple cysts in the kidneys. It is often caused by pathogenic mutations in PKD1 and PKD2 genes that encode polycystin proteins. Although the molecular mechanisms for cystogenesis are not established, concurrent inactivating germline and somatic mutations in PKD1 and PKD2 have been previously observed in renal tubular epithelium (RTE). METHODS To further investigate the cellular recessive mechanism of cystogenesis in RTE, we conducted whole-genome DNA sequencing analysis to identify germline variants and somatic alterations in RTE of 90 unique kidney cysts obtained during nephrectomy from 24 unrelated participants. RESULTS Kidney cysts were overall genomically stable, with low burdens of somatic short mutations or large-scale structural alterations. Pathogenic somatic "second hit" alterations disrupting PKD1 or PKD2 were identified in 93% of the cysts. Of these, 77% of cysts acquired short mutations in PKD1 or PKD2 ; specifically, 60% resulted in protein truncations (nonsense, frameshift, or splice site) and 17% caused non-truncating mutations (missense, in-frame insertions, or deletions). Another 18% of cysts acquired somatic chromosomal loss of heterozygosity (LOH) events encompassing PKD1 or PKD2 ranging from 2.6 to 81.3 Mb. 14% of these cysts harbored copy number neutral LOH events, while the other 3% had hemizygous chromosomal deletions. LOH events frequently occurred at chromosomal fragile sites, or in regions comprising chromosome microdeletion diseases/syndromes. Almost all somatic "second hit" alterations occurred at the same germline mutated PKD1/2 gene. CONCLUSIONS These findings further support a cellular recessive mechanism for cystogenesis in ADPKD primarily caused by inactivating germline and somatic variants of PKD1 or PKD2 genes in kidney cyst epithelium.
Collapse
Affiliation(s)
- Zhengmao Zhang
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Hanwen Bai
- Vertex Pharmaceuticals Inc., Boston, Massachusetts
| | - Jon Blumenfeld
- Department of Medicine, Weill Cornell Medicine, New York, New York
- The Rogosin Institute, New York, New York
| | - Andrew B. Ramnauth
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Irina Barash
- Department of Medicine, Weill Cornell Medicine, New York, New York
- The Rogosin Institute, New York, New York
| | - Martin Prince
- Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Adrian Y. Tan
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Alber Michaeel
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Genyan Liu
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | | | - Lior Rennert
- Department of Public Health Sciences, Clemson University, Clemson, South Carolina
| | | | - Karen Larbi
- Vertex Pharmaceuticals Inc., Oxford, United Kingdom
| | | | - Steven P. Salvatore
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Brian D. Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Sandip Kapur
- Department of Surgery, Weill Cornell Medicine, New York, New York
| | - Hanna Rennert
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| |
Collapse
|
20
|
Nielsen ML, Mundt MC, Lildballe DL, Rasmussen M, Sunde L, Torres VE, Harris PC, Birn H. Functional megalin is expressed in renal cysts in a mouse model of adult polycystic kidney disease. Clin Kidney J 2021; 14:2420-2427. [PMID: 34754438 PMCID: PMC8572980 DOI: 10.1093/ckj/sfab088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/06/2021] [Indexed: 12/14/2022] Open
Abstract
Background Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the progressive growth of cysts and a decline of renal function. The clinical feasibility of the number of potential disease-modifying drugs is limited by systemic adverse effects. We hypothesize that megalin, a multiligand endocytic receptor expressed in the proximal tubule, may be used to facilitate drug uptake into cysts, thereby allowing for greater efficacy and fewer side effects. Methods The cyst expression of various tubular markers, including megalin and aquaporin 2 (AQP2), was analysed by immunohistochemistry (IHC) of kidney sections from the ADPKD mouse model (PKD1RC/RC) at different post-natal ages. The endocytic function of megalin in cysts was examined by IHC of kidney tissue from mice injected with the megalin ligand aprotinin. Results Cyst lining epithelial cells expressing megalin were observed at all ages; however, the proportion decreased with age. Concomitantly, an increasing proportion of cysts revealed expression of AQP2, partial expression of megalin and/or AQP2 or no expression of the examined markers. Endocytic uptake of aprotinin was evident in megalin-positive cysts, but only in those that remained connected to the renal tubular system. Conclusions Megalin-expressing cysts were observed at all ages, but the proportion decreased with age, possibly due to a switch in tubular origin, a merging of cysts of different tubular origin and/or a change in the expression pattern of cyst lining cells. Megalin expressed in cysts was functional, suggesting that megalin-mediated endocytosis is a potential mechanism for drug targeting in ADPKD if initiated early in the disease.
Collapse
Affiliation(s)
| | - Mia C Mundt
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Dorte L Lildballe
- Department of Molecular Medine, Aarhus University Hospital, Aarhus N, Denmark
| | - Maria Rasmussen
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, Vejle, Denmark
| | - Lone Sunde
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| | - Vicente E Torres
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Peter C Harris
- Department of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| | - Henrik Birn
- Department of Biomedicine, Aarhus University, Aarhus C, Denmark
| |
Collapse
|
21
|
Hu H, Zhang J, Qiu W, Liang C, Li C, Wei T, Feng Z, Guo Q, Yang K, Liu Z. Comprehensive strategy improves the genetic diagnosis of different polycystic kidney diseases. J Cell Mol Med 2021; 25:6318-6332. [PMID: 34032358 PMCID: PMC8256360 DOI: 10.1111/jcmm.16608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 04/26/2021] [Indexed: 12/16/2022] Open
Abstract
Polycystic kidney disease (PKD) is known to occur in three main forms, namely autosomal dominant PKD (ADPKD), autosomal recessive PKD (ARPKD) and syndromic PKD (SPKD), based on the clinical manifestations and genetic causes, which are diagnosable from the embryo stage to the later stages of life. Selection of the genetic test for the individuals with diagnostic imaging reports of cystic kidneys without a family history of the disease continues to be a challenge in clinical practice. With the objective of maintaining a limit on the time and medical cost of the procedure, a practical strategy for genotyping and targeted validation to resolve cystogene variations was developed in our clinical laboratory, which combined the techniques of whole-exome sequencing (WES), Long-range PCR (LR-PCR), Sanger sequencing and multiplex ligation-dependent probe amplification (MLPA) to work in a stepwise approach. In this context, twenty-six families with renal polycystic disorders were enrolled in the present study. Thirty-two variants involving four ciliary genes (PKD1, PKHD1, TMEM67 and TMEM107) were identified and verified in 23 families (88.5%, 23/26), which expanded the variant spectrum by 16 novel variants. Pathogenic variations in five foetuses of six families diagnosed with PKD were identified using prenatal ultrasound imaging. Constitutional biallelic and digenic variations constituted the pathogenic patterns in these foetuses. The preliminary clinical data highlighted that the WES + LR PCR-based workflow followed in the present study is efficient in detecting divergent variations in PKD. The biallelic and digenic mutations were revealed as the main pathogenic patterns in the foetuses with PKD.
Collapse
Affiliation(s)
- Hua‐Ying Hu
- Department of OphthalmologyXiang'an Hospital of Xiamen UniversityFujian Provincial Key Laboratory of Ophthalmology and Visual ScienceSchool of Medicine, Xiamen UniversityFujian Engineering and Research Center of Eye Regenerative MedicineEye Institute of Xiamen UniversityXiamenChina
- Jiaen Genetics LaboratoryBeijing Jiaen HospitalBeijingChina
| | - Jing Zhang
- Prenatal Diagnosis CenterShijiazhuang Obstetrics and Gynecology HospitalHebeiChina
| | - Wei Qiu
- Department of UrologyBeijing Friendship HospitalCapital Medical UniversityBeijingChina
| | - Chao Liang
- Department of Pediatric OrthopedicsShijiazhuang Obstetrics and Gynecology HospitalHebeiChina
| | - Cun‐Xi Li
- Jiaen Genetics LaboratoryBeijing Jiaen HospitalBeijingChina
| | - Tian‐Ying Wei
- Jiaen Genetics LaboratoryBeijing Jiaen HospitalBeijingChina
| | - Zhan‐Ke Feng
- Jiaen Genetics LaboratoryBeijing Jiaen HospitalBeijingChina
| | - Qing Guo
- Prenatal Diagnosis CenterShijiazhuang Obstetrics and Gynecology HospitalHebeiChina
| | - Kai Yang
- Prenatal Diagnosis CenterBeijing Obstetrics and Gynecology Hospital, Capital Medical UniversityBeijingChina
| | - Zu‐Guo Liu
- Department of OphthalmologyXiang'an Hospital of Xiamen UniversityFujian Provincial Key Laboratory of Ophthalmology and Visual ScienceSchool of Medicine, Xiamen UniversityFujian Engineering and Research Center of Eye Regenerative MedicineEye Institute of Xiamen UniversityXiamenChina
| |
Collapse
|
22
|
Jung SH, You JE, Choi SW, Kang KS, Cho JY, Lyu J, Kim PH. Polycystin-1 Enhances Stemmness Potential of Umbilical Cord Blood-Derived Mesenchymal Stem Cells. Int J Mol Sci 2021; 22:ijms22094868. [PMID: 34064452 PMCID: PMC8125233 DOI: 10.3390/ijms22094868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/28/2021] [Accepted: 05/01/2021] [Indexed: 01/01/2023] Open
Abstract
Polycystic Kidney Disease (PKD) is a disorder that affects the kidneys and other organs, and its major forms are encoded by polycystin-1 (PC1) and polycystin-2 (PC2), as PKD1 and PKD2. It is located sandwiched inside and outside cell membranes and interacts with other cells. This protein is most active in kidney cells before birth, and PC1 and PC2 work together to help regulate cell proliferation, cell migration, and interactions with other cells. The molecular relationship and the function between PKD1 and cancer is well known, such as increased or decreased cell proliferation and promoting or suppressing cell migration depending on the cancer cell type specifically. However, its function in stem cells has not been revealed. Therefore, in this study, we investigated the biological function of PC1 and umbilical cord blood-derived mesenchymal stem cell (UCB-MSC). Furthermore, we assessed how it affects cell migration, proliferation, and the viability of cells when expressed in the PKD1 gene. In addition, we confirmed in an ex vivo artificial tooth model generated by the three-dimension printing technique that the ability to differentiate into osteocytes improved according to the expression level of the stemness markers when PKD1 was expressed. This study is the first report to examine the biological function of PKD1 in UCB-MSC. This gene may be capable of enhancing differentiation ability and maintaining long-term stemness for the therapeutic use of stem cells.
Collapse
Affiliation(s)
- Se-Hwa Jung
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (S.-H.J.); (J.-E.Y.)
| | - Ji-Eun You
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (S.-H.J.); (J.-E.Y.)
| | - Soon-Won Choi
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.-W.C.); (K.-S.K.)
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea; (S.-W.C.); (K.-S.K.)
| | - Je-Yeol Cho
- Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea;
| | - Jungmook Lyu
- Myung-Gok Eye Research Institute, Department of Medical Science, Konyang University, Daejeon 320-832, Korea;
| | - Pyung-Hwan Kim
- Department of Biomedical Laboratory Science, Konyang University, Daejeon 35365, Korea; (S.-H.J.); (J.-E.Y.)
- Correspondence: ; Tel.: +82-42-600-8436; Fax: +82-42-600-8408
| |
Collapse
|
23
|
Morozov D, Parvin N, Charlton JR, Bennett KM. Mapping kidney tubule diameter ex vivo by diffusion MRI. Am J Physiol Renal Physiol 2021; 320:F934-F946. [PMID: 33719573 DOI: 10.1152/ajprenal.00369.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Tubular pathologies are a common feature of kidney disease. Current metrics to assess kidney health, in vivo or in transplant, are generally based on urinary or serum biomarkers and pathological findings from kidney biopsies. Biopsies, usually taken from the kidney cortex, are invasive and prone to sampling error. Tools to directly and noninvasively measure tubular pathology could provide a new approach to assess kidney health. This study used diffusion magnetic resonance imaging (dMRI) as a noninvasive tool to measure the size of the tubular lumen in ex vivo, perfused kidneys. We first used Monte Carlo simulations to demonstrate that dMRI is sensitive to restricted tissue water diffusion at the scale of the kidney tubule. We applied dMRI and biophysical modeling to examine the distribution of tubular diameters in ex vivo, fixed kidneys from mice, rats, and a human donor. The biophysical model to fit the dMRI signal was based on a superposition of freely diffusing water and water diffusing inside infinitely long cylinders of different diameters. Tubular diameters measured by dMRI were within 10% of those measured by histology within the same tissue. Finally, we applied dMRI to investigate kidney pathology in a mouse model of folic-acid-induced acute kidney injury. dMRI detected heterogeneity in the distribution of tubules within the kidney cortex of mice with acute kidney injury compared with control mice. We conclude that dMRI can be used to measure the distribution of tubule diameters in the kidney cortex ex vivo and that dMRI may provide a new noninvasive biomarker of tubular pathology.NEW & NOTEWORTHY Tubular pathologies are a common feature of kidney disease. Current metrics to assess kidney health, in vivo or in transplant, are generally based on urinary or serum biomarkers and pathological findings from kidney biopsies. Diffusion MRI can be used to measure the distribution of tubule diameters in the kidney cortex ex vivo and may provide a new noninvasive biomarker of tubular pathology.
Collapse
Affiliation(s)
- Darya Morozov
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | - Neda Parvin
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| | | | - Kevin M Bennett
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
24
|
Clinical genetic diagnostics in Danish autosomal dominant polycystic kidney disease patients reveal possible founder variants. Eur J Med Genet 2021; 64:104183. [PMID: 33639313 DOI: 10.1016/j.ejmg.2021.104183] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/04/2021] [Accepted: 02/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is the most common heritable kidney disease. ADPKD leads to cysts, kidney enlargement and end-stage renal disease. ADPKD is mainly caused by variants in PKD1 and PKD2, with truncating PKD1 variants causing the most severe phenotype. This study aimed to characterize variants in Danish patients referred for screening of genes related to cystic kidney disease. METHODS 147 families were analysed for variants in PKD1, PKD2 and GANAB using next generation sequencing and multiplex ligation-dependent probe amplification. If a variant was identified, relatives were analysed for the specific variant using Sanger sequencing. RESULTS A pathogenic or possibly pathogenic variant was identified in 87% (103/118) of patients suspected to suffer from ADPKD, according to the requisition form. In total, 112 pathogenic or possibly pathogenic variants were observed, of which 94 were unique; 74 (79%) in PKD1 and 20 (21%) in PKD2, while 41 variants were novel. No variants in GANAB were observed. Ten recurrent variants were observed in 26 (26%) families. These were either PKD2 variants (N = 6) or non-truncating PKD1 variants (N = 4). Five of these were likely founder variants. CONCLUSIONS The distribution of pathogenic or possibly pathogenic variants in the Danish ADPKD population is similar to that in other populations, except that recurrent truncating PKD1 variants appear to be rare, i.e. founder variants tend to be variant types associated with a mild phenotype. Patients with a mild phenotype may remain undiagnosed, consequently the frequency of founder variants and prevalence of ADPKD may be underestimated.
Collapse
|
25
|
Histomorphology and Immunohistochemistry of a Congenital Nephromegaly Demonstrate Concurrent Features of Heritable and Acquired Cystic Nephropathies in a Girgentana Goat ( Capra falconeri). Case Rep Vet Med 2021; 2021:8749158. [PMID: 33532110 PMCID: PMC7837792 DOI: 10.1155/2021/8749158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 12/09/2020] [Accepted: 12/31/2020] [Indexed: 11/17/2022] Open
Abstract
Polycystic kidney diseases (PKD) represent frequent congenital and adult nephropathies in humans and domestic animals. This report illustrates an uncommon state of congenital PKD in a girgentana goat (Capra falconeri). A stillborn female goat kid was submitted for postmortem examination and underwent macroscopic and microscopic examination. The kidneys showed a bilateral nephromegaly and a perpendicular polycystic altered texture of the renal parenchyma. Renal tissue sections were comprehensively investigated by histopathology (overview and special stains), immunohistochemistry (CD10, CD117, pan-cytokeratin, cytokeratin 7, E-cadherin, Pax2, Pax8, and vimentin), and electron microscopy (SEM, TEM). Histopathology of renal tissue sections revealed polycystic alterations of the renal parenchyma as well as conspicuous polypoid proliferates/projections of the renal tubular epithelium, which showed clear cell characteristics. Furthermore, epithelial projections were indicative for epithelio-mesenchymal-transition, cellular depolarization, and strong expression of differentiation markers Pax2, Pax8, and CD10. Ultrastructural morphology of the projections was characterized by numerous diffusely distributed, demarked round cytoplasmic structures and several apico-lateral differentiations. Additionally, hepatic malformations comprising biliary duct proliferation with saccular dilation and bridging fibrosis were observed. Notably, this report describes the first case of a congenital cystic nephropathy with overlapping features of heritable and acquired nephropathies in any species. Epithelio-mesenchymal-transition and altered cadherin expression seem to be crucial components of a suspected pathomechanism during cystogenesis.
Collapse
|
26
|
Connor KL, Teenan O, Cairns C, Banwell V, Thomas RA, Rodor J, Finnie S, Pius R, Tannahill GM, Sahni V, Savage CO, Hughes J, Harrison EM, Henderson RB, Marson LP, Conway BR, Wigmore SJ, Denby L. Identifying cell-enriched miRNAs in kidney injury and repair. JCI Insight 2020; 5:140399. [PMID: 33328386 PMCID: PMC7819746 DOI: 10.1172/jci.insight.140399] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/06/2020] [Indexed: 12/14/2022] Open
Abstract
Small noncoding RNAs, miRNAs (miRNAs), are emerging as important modulators in the pathogenesis of kidney disease, with potential as biomarkers of kidney disease onset, progression, or therapeutic efficacy. Bulk tissue small RNA-sequencing (sRNA-Seq) and microarrays are widely used to identify dysregulated miRNA expression but are limited by the lack of precision regarding the cellular origin of the miRNA. In this study, we performed cell-specific sRNA-Seq on tubular cells, endothelial cells, PDGFR-β+ cells, and macrophages isolated from injured and repairing kidneys in the murine reversible unilateral ureteric obstruction model. We devised an unbiased bioinformatics pipeline to define the miRNA enrichment within these cell populations, constructing a miRNA catalog of injury and repair. Our analysis revealed that a significant proportion of cell-specific miRNAs in healthy animals were no longer specific following injury. We then applied this knowledge of the relative cell specificity of miRNAs to deconvolute bulk miRNA expression profiles in the renal cortex in murine models and human kidney disease. Finally, we used our data-driven approach to rationally select macrophage-enriched miR-16-5p and miR-18a-5p and demonstrate that they are promising urinary biomarkers of acute kidney injury in renal transplant recipients.
Collapse
Affiliation(s)
- Katie L Connor
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Transplant Unit, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.,Centre for Inflammation Research and
| | - Oliver Teenan
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Carolynn Cairns
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Victoria Banwell
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Transplant Unit, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.,Centre for Inflammation Research and
| | - Rachel Ab Thomas
- Edinburgh Transplant Unit, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.,Centre for Inflammation Research and
| | - Julie Rodor
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Sarah Finnie
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Riinu Pius
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Vishal Sahni
- Medicines Research Centre, GlaxoSmithKline, Stevenage, United Kingdom
| | | | | | - Ewen M Harrison
- Edinburgh Transplant Unit, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.,Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, United Kingdom
| | | | - Lorna P Marson
- Edinburgh Transplant Unit, Edinburgh Royal Infirmary, Edinburgh, United Kingdom.,Centre for Inflammation Research and
| | - Bryan R Conway
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen J Wigmore
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom.,Edinburgh Transplant Unit, Edinburgh Royal Infirmary, Edinburgh, United Kingdom
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Wang H, Dai S, Zhang J, Li Y, Gan Y, Lu T, Zhu Y, Wu J, Lin N, Tang F, Luo J. Analysis of mutations in six Chinese families with autosomal dominant polycystic kidney disease. Am J Transl Res 2020; 12:8123-8136. [PMID: 33437386 PMCID: PMC7791523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/31/2020] [Indexed: 06/12/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the common hereditary kidney disease, resulting from mutations in polycystic kidney disease 1 (PKD1) and polycystic kidney disease 2 (PKD2). Clinical data and genetic features of six Chinese families including ADPKD patients were analyzed via Next generation sequencing (NGS), Sanger sequencing, and multiplex ligation-dependent probe amplification. In family A, the proband (II5) with polycystic kidney (PK), hypertension, left ventricular hypertrophy, and valvular heart disease exhibited a heterozygous nonsense mutation, c.5086C>T (p.Gln1696Ter), in PKD1 (NM_001009944). In family B, the proband (II3) with PK, polycystic liver (PL), hypertension, hypertrophy of the left ventricle and septum, valvular heart disease, chronic kidney disease (CKD) stage 5, bilateral renal calculi, and right inguinal hernia exhibited a heterozygous missense mutation, c.6695T>C (p.Phe2232Ser), in PKD1. In family C, the proband (III1) with PK, PL, seminal vesicle cyst, hypertension, CKD stage 3, hypertrophy of the left ventricle and septum, and valvular heart disease harbored a heterozygous nonsense mutation, c.662T>G (p.Leu221Ter), in PKD2 (NM_000297). In family D, the proband (III3) with PK, hypertension, and CKD stage 5 harbored a heterozygous missense mutation, c.8311G>A (p.Glu2771Lys), in PKD1. In family E, the proband (II1) with PK, PL, hypertension, and CKD stage 5 exhibited a heterozygous deletion mutation, exon15-22, in PKD1. In family F, the proband (II2) with PK, PL, CKD stage 3, hypertension, thickened interventricular septum, and valvular heart disease carried a heterozygous missense mutation, c.1649A>G (p.His550Arg), in PKD2. Thus, three novel mutation sites which are responsible for ADPKD were discovered in this study.
Collapse
Affiliation(s)
- Hanlu Wang
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Sen Dai
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Jianhui Zhang
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Yi Li
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical UniversityNanjing 210009, China
| | - Yumian Gan
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Tao Lu
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Yaobin Zhu
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Fujian Medical UniversityFuzhou 350001, China
| | - Jiabin Wu
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Ning Lin
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Faqiang Tang
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| | - Jiewei Luo
- Shengli Clinical Medical College of Fujian Medical UniversityFuzhou 350001, China
- Fujian Provincial HospitalFuzhou 350001, China
| |
Collapse
|
28
|
Saini AK, Saini R, Singh S. Autosomal dominant polycystic kidney disease and pioglitazone for its therapy: a comprehensive review with an emphasis on the molecular pathogenesis and pharmacological aspects. Mol Med 2020; 26:128. [PMID: 33308138 PMCID: PMC7731470 DOI: 10.1186/s10020-020-00246-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is an inherited chronic kidney disorder (CKD) that is characterized by the development of numerous fluid-filled cysts in kidneys. It is caused either due to the mutations in the PKD1 or PKD2 gene that encodes polycystin-1 and polycystin-2, respectively. This condition progresses into end-stage renal disorder if the renal or extra-renal clinical manifestations remain untreated. Several clinical trials with a variety of drugs have failed, and the only Food and Drugs Administration (FDA) approved drug to treat ADPKD to date is tolvaptan that works by antagonizing the vasopressin-2 receptor (V2R). The pathology of ADPKD is complex and involves the malfunction of different signaling pathways like cAMP, Hedgehog, and MAPK/ERK pathway owing to the mutated product that is polycystin-1 or 2. A measured yet substantial number of preclinical studies have found pioglitazone to decrease the cystic burden and improve the renal function in ADPKD. The peroxisome proliferator-activated receptor-gamma is found on the epithelial cells of renal collecting tubule and when it gets agonized by pioglitazone, confers efficacy in ADPKD treatment through multiple mechanisms. There is only one clinical trial (ongoing) wherein it is being assessed for its benefits and risk in patients with ADPKD, and is expected to get approval from the regulatory body owing to its promising therapeutic effects. This article would encompass the updated information on the epidemiology, pathophysiology of ADPKD, different mechanisms of action of pioglitazone in the treatment of ADPKD with preclinical and clinical shreds of evidence, and related safety updates.
Collapse
Affiliation(s)
- Aryendu Kumar Saini
- Department of Pharmacy, Chaudhary Sughar Singh College of Pharmacy, Etawah, Uttar Pradesh, India.
| | - Rakesh Saini
- Department of Pharmacy, Chaudhary Sughar Singh College of Pharmacy, Etawah, Uttar Pradesh, India
| | - Shubham Singh
- Department of Pharmacy, Shri Ram Lakhan Tiwari College of Pharmacy, Etawah, Uttar Pradesh, India
| |
Collapse
|
29
|
Viola JM, Porter CM, Gupta A, Alibekova M, Prahl LS, Hughes AJ. Guiding Cell Network Assembly using Shape-Morphing Hydrogels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2002195. [PMID: 32578300 PMCID: PMC7950730 DOI: 10.1002/adma.202002195] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/30/2020] [Indexed: 05/11/2023]
Abstract
Forces and relative movement between cells and extracellular matrix (ECM) are crucial to the self-organization of tissues during development. However, the spatial range over which these dynamics can be controlled in engineering approaches is limited, impeding progress toward the construction of large, structurally mature tissues. Herein, shape-morphing materials called "kinomorphs" that rationally control the shape and size of multicellular networks are described. Kinomorphs are sheets of ECM that change their shape, size, and density depending on patterns of cell contractility within them. It is shown that these changes can manipulate structure-forming behaviors of epithelial cells in many spatial locations at once. Kinomorphs are built using a new photolithographic technology to pattern single cells into ECM sheets that are >10× larger than previously described. These patterns are designed to partially mimic the branch geometry of the embryonic kidney epithelial network. Origami-inspired simulations are then used to predict changes in kinomorph shapes. Last, kinomorph dynamics are shown to provide a centimeter-scale program that sets specific spatial locations in which ≈50 µm-diameter epithelial tubules form by cell coalescence and structural maturation. The kinomorphs may significantly advance organ-scale tissue construction by extending the spatial range of cell self-organization in emerging model systems such as organoids.
Collapse
Affiliation(s)
- John M Viola
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Catherine M Porter
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Ananya Gupta
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Mariia Alibekova
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Louis S Prahl
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Alex J Hughes
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
30
|
An Overview of In Vivo and In Vitro Models for Autosomal Dominant Polycystic Kidney Disease: A Journey from 3D-Cysts to Mini-Pigs. Int J Mol Sci 2020; 21:ijms21124537. [PMID: 32630605 PMCID: PMC7352572 DOI: 10.3390/ijms21124537] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/24/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inheritable cause of end stage renal disease and, as of today, only a single moderately effective treatment is available for patients. Even though ADPKD research has made huge progress over the last decades, the precise disease mechanisms remain elusive. However, a wide variety of cellular and animal models have been developed to decipher the pathophysiological mechanisms and related pathways underlying the disease. As none of these models perfectly recapitulates the complexity of the human disease, the aim of this review is to give an overview of the main tools currently available to ADPKD researchers, as well as their main advantages and limitations.
Collapse
|
31
|
Womble MA, Lewbart GA, Shive HR. Pathologic Lesions of the Budgett Frog ( Lepidobatrachus laevis), an Emerging Laboratory Animal Model. Comp Med 2020; 70:239-247. [PMID: 32234112 DOI: 10.30802/aalas-cm-19-000071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lepidobatrachus laevis, commonly called the Budgett frog, is a member of the horned frog family (Ceratophryidae), which has become increasingly popular among amphibian hobbyists. L. laevis is also used in biologic research on embryonic development, providing a novel model species for the study of organogenesis, regeneration, evolution, and biologic scaling. However, little scientific literature details disease processes or histologic lesions in this species. Our objective was to describe spontaneous pathologic lesions in L. laevis to identify disease phenotypes. We performed a retrospective analysis of 14 captive L. laevis frogs (wild-caught and captive-bred), necropsied at the NC State University College of Veterinary Medicine between 2008 and 2018. The majority of frogs exhibited renal changes, including varying combinations of tubular epithelial binucleation, karyomegaly, and cytoplasmic vacuolation; polycystic kidney disease; and renal carcinoma. Many of the renal changes are reminiscent of a condition described in Japanese (Bufo japonicus) and Chinese (Bufo raddei) toad hybrids that progresses from tubular epithelial atypia and tubular dilation to polycystic kidney disease to renal carcinoma. A second common finding was variably sized, randomly distributed bile duct clusters (biliary proliferation). Other noteworthy findings included regional or generalized edema, intestinal adenocarcinoma, aspiration pneumonia, and parasitism. This retrospective analysis is the first description of histologic lesions identified in captive L. laevis populations, providing new insight into spontaneous disease processes occurring in this species for use in disease diagnosis and clinical management.
Collapse
Affiliation(s)
- Mandy A Womble
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina
| | - Gregory A Lewbart
- Department of Clinical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina
| | - Heather R Shive
- Department of Population Health and Pathobiology, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina;,
| |
Collapse
|
32
|
Takebayashi-Suzuki K, Suzuki A. Intracellular Communication among Morphogen Signaling Pathways during Vertebrate Body Plan Formation. Genes (Basel) 2020; 11:E341. [PMID: 32213808 PMCID: PMC7141137 DOI: 10.3390/genes11030341] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/18/2020] [Accepted: 03/19/2020] [Indexed: 12/25/2022] Open
Abstract
During embryonic development in vertebrates, morphogens play an important role in cell fate determination and morphogenesis. Bone morphogenetic proteins (BMPs) belonging to the transforming growth factor-β (TGF-β) family control the dorsal-ventral (DV) patterning of embryos, whereas other morphogens such as fibroblast growth factor (FGF), Wnt family members, and retinoic acid (RA) regulate the formation of the anterior-posterior (AP) axis. Activation of morphogen signaling results in changes in the expression of target genes including transcription factors that direct cell fate along the body axes. To ensure the correct establishment of the body plan, the processes of DV and AP axis formation must be linked and coordinately regulated by a fine-tuning of morphogen signaling. In this review, we focus on the interplay of various intracellular regulatory mechanisms and discuss how communication among morphogen signaling pathways modulates body axis formation in vertebrate embryos.
Collapse
Affiliation(s)
- Kimiko Takebayashi-Suzuki
- Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima 739-8526, Japan
| | - Atsushi Suzuki
- Graduate School of Integrated Sciences for Life, Amphibian Research Center, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, Hiroshima, 739-8526, Japan
| |
Collapse
|
33
|
Tutunea-Fatan E, Lee JC, Denker BM, Gunaratnam L. Heterotrimeric Gα 12/13 proteins in kidney injury and disease. Am J Physiol Renal Physiol 2020; 318:F660-F672. [PMID: 31984793 DOI: 10.1152/ajprenal.00453.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gα12 and Gα13 are ubiquitous members of the heterotrimeric guanine nucleotide-binding protein (G protein) family that play central and integrative roles in the regulation of signal transduction cascades within various cell types in the kidney. Gα12/Gα13 proteins enable the kidney to adapt to an ever-changing environment by transducing stimuli from cell surface receptors and accessory proteins to effector systems. Therefore, perturbations in Gα12/Gα13 levels or their activity can contribute to the pathogenesis of various renal diseases, including renal cancer. This review will highlight and discuss the complex and expanding roles of Gα12/Gα13 proteins on distinct renal pathologies, with emphasis on more recently reported findings. Deciphering how the different Gα12/Gα13 interaction networks participate in the onset and development of renal diseases may lead to the discovery of new therapeutic strategies.
Collapse
Affiliation(s)
- Elena Tutunea-Fatan
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada
| | - Jasper C Lee
- Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada
| | - Bradley M Denker
- Beth Israel Deaconess Medical Center and Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Lakshman Gunaratnam
- Matthew Mailing Centre for Translational Transplant Studies, Lawson Health Research Institute, London, Ontario, Canada.,Department of Microbiology and Immunology, University of Western Ontario, London, Ontario, Canada.,Division of Nephrology, Department of Medicine, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
34
|
Papavassiliou KA, Zoi I, Gargalionis AN, Koutsilieris M. Polycystin-1 affects cancer cell behaviour and interacts with mTOR and Jak signalling pathways in cancer cell lines. J Cell Mol Med 2019; 23:6215-6227. [PMID: 31251475 PMCID: PMC6714176 DOI: 10.1111/jcmm.14506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 01/28/2023] Open
Abstract
Polycystic Kidney Disease (PKD), which is attributable to mutations in the PKD1 and PKD2 genes encoding polycystin‐1 (PC1) and polycystin‐2 (PC2) respectively, shares common cellular defects with cancer, such as uncontrolled cell proliferation, abnormal differentiation and increased apoptosis. Interestingly, PC1 regulates many signalling pathways including Jak/STAT, mTOR, Wnt, AP‐1 and calcineurin‐NFAT which are also used by cancer cells for sending signals that will allow them to acquire and maintain malignant phenotypes. Nevertheless, the molecular relationship between polycystins and cancer is unknown. In this study, we investigated the role of PC1 in cancer biology using glioblastoma (GOS3), prostate (PC3), breast (MCF7), lung (A549) and colorectal (HT29) cancer cell lines. Our in vitro results propose that PC1 promotes cell migration in GOS3 cells and suppresses cell migration in A549 cells. In addition, PC1 enhances cell proliferation in GOS3 cells but inhibits it in MCF7, A549 and HT29 cells. We also found that PC1 up‐regulates mTOR signalling and down‐regulates Jak signalling in GOS3 cells, while it up‐regulates mTOR signalling in PC3 and HT29 cells. Together, our study suggests that PC1 modulates cell proliferation and migration and interacts with mTOR and Jak signalling pathways in different cancer cell lines. Understanding the molecular details of how polycystins are associated with cancer may lead to the identification of new players in this devastating disease.
Collapse
Affiliation(s)
- Kostas A Papavassiliou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ilianna Zoi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios N Gargalionis
- Department of Biopathology, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
35
|
Mamone G, Carollo V, Cortis K, Aquilina S, Liotta R, Miraglia R. Magnetic resonance imaging of fibropolycystic liver disease: the spectrum of ductal plate malformations. Abdom Radiol (NY) 2019; 44:2156-2171. [PMID: 30852632 DOI: 10.1007/s00261-019-01966-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Fibropolycystic liver diseases, also known as ductal plate malformations, are a group of associated congenital disorders resulting from abnormal development of the biliary ductal system. These disorders include congenital hepatic fibrosis, biliary hamartomas, polycystic liver disease, choledochal cysts and Caroli disease. Recently, it has been thought to include biliary atresia in this group of diseases, because ductal plate malformations could be implicated in the pathogenesis of this disease. Concomitant associated renal anomalies can also be present, such as autosomal recessive polycystic kidney disease (ARPKD), medullary sponge kidney and nephronophthisis. These disorders can be clinically silent or can cause abnormalities such as cholangitis, portal hypertension, gastrointestinal bleeding and infections. The different types of ductal plate malformations show typical findings at magnetic resonance (MR) imaging. A clear knowledge of the embryology and pathogenesis of the ductal plate plays a pivotal role to understand the characteristic imaging appearances of these complex diseases. Awareness of these MR imaging findings is central to the detecting and differentiating between various fibropolycystic liver diseases and is important to direct appropriate clinical management and prevent misdiagnosis.
Collapse
Affiliation(s)
- Giuseppe Mamone
- Radiology Unit, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127, Palermo, Italy.
| | - Vincenzo Carollo
- Radiology Unit, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127, Palermo, Italy
| | - Kelvin Cortis
- Department of Medical Imaging, Mater Dei Hospital, Msida, MSD 2090, Malta
| | - Sarah Aquilina
- Department of Medical Imaging, Mater Dei Hospital, Msida, MSD 2090, Malta
| | - Rosa Liotta
- Pathology Unit, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127, Palermo, Italy
| | - Roberto Miraglia
- Radiology Unit, Department of Diagnostic and Therapeutic Services, IRCCS ISMETT (Mediterranean Institute for Transplantation and Advanced Specialized Therapies), Via Tricomi 5, 90127, Palermo, Italy
| |
Collapse
|
36
|
Potts JW, Mousa SA. Recent advances in management of autosomal-dominant polycystic kidney disease. Am J Health Syst Pharm 2019; 74:1959-1968. [PMID: 29167138 DOI: 10.2146/ajhp160886] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Promising developments in the search for effective pharmacotherapies for autosomal-dominant polycystic kidney disease (ADPKD) are reviewed. SUMMARY The formation and development of cysts characteristic of ADPKD result in inexorable renal and extrarenal manifestations that give rise to more rapid disease progression and more widespread complications than are seen with other forms of chronic kidney disease. To date, no agent has gained Food and Drug Administration marketing approval for use in patients with ADPKD, complicating efforts to meet the medical needs of this population. Although definitive ultrasonographic diagnostic strategies are available, molecular screening approaches lack sufficient evidence and patient outcomes data to support broad clinical application. Recently completed and ongoing clinical trials point to a number of encouraging platforms for evidence-based ADPKD management. Tolvaptan therapy significantly improved cyst burden and slowed disease progression among patients with early-stage ADPKD in a large-scale trial, while somatostatin therapies may also be useful in halting disease progression and managing comorbid polycystic liver disease. Stem cell research and nanomedicine might represent novel approaches to gaining comprehensive insights on ADPKD and, ultimately, to targeting the disease's origins, thereby making restoration of kidney function possible. CONCLUSION A number of pharmacotherapy approaches to ADPKD management show promise but are unlikely to be curative, fueling interest among researchers in finding new applications for nanomedicine and stem cell technologies that can slow ADPKD progression and better control complications of the disease.
Collapse
Affiliation(s)
- Jacob W Potts
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY
| | - Shaker A Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY
| |
Collapse
|
37
|
Kokil P, Sudharson S. Automatic Detection of Renal Abnormalities by Off-the-shelf CNN Features. ACTA ACUST UNITED AC 2019. [DOI: 10.1080/09747338.2019.1613936] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Priyanka Kokil
- Department of Electronics and Communication Engineering, Indian Institute of Information Technology, Design and Manufacturing, Kancheepuram, 600127 Chennai, India
| | - S. Sudharson
- Department of Electronics and Communication Engineering, Indian Institute of Information Technology, Design and Manufacturing, Kancheepuram, 600127 Chennai, India
| |
Collapse
|
38
|
Parnell SC, Magenheimer BS, Maser RL, Pavlov TS, Havens MA, Hastings ML, Jackson SF, Ward CJ, Peterson KR, Staruschenko A, Calvet JP. A mutation affecting polycystin-1 mediated heterotrimeric G-protein signaling causes PKD. Hum Mol Genet 2019; 27:3313-3324. [PMID: 29931260 PMCID: PMC6140781 DOI: 10.1093/hmg/ddy223] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 06/05/2018] [Indexed: 12/16/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the growth of renal cysts that ultimately destroy kidney function. Mutations in the PKD1 and PKD2 genes cause ADPKD. Their protein products, polycystin-1 (PC1) and polycystin-2 (PC2) have been proposed to form a calcium-permeable receptor-channel complex; however the mechanisms by which they function are almost completely unknown. Most mutations in PKD1 are truncating loss-of-function mutations or affect protein biogenesis, trafficking or stability and reveal very little about the intrinsic biochemical properties or cellular functions of PC1. An ADPKD patient mutation (L4132Δ or ΔL), resulting in a single amino acid deletion in a putative G-protein binding region of the PC1 C-terminal cytosolic tail, was found to significantly decrease PC1-stimulated, G-protein-dependent signaling in transient transfection assays. Pkd1ΔL/ΔL mice were embryo-lethal suggesting that ΔL is a functionally null mutation. Kidney-specific Pkd1ΔL/cond mice were born but developed severe, postnatal cystic disease. PC1ΔL protein expression levels and maturation were comparable to those of wild type PC1, and PC1ΔL protein showed cell surface localization. Expression of PC1ΔL and PC2 complexes in transfected CHO cells failed to support PC2 channel activity, suggesting that the role of PC1 is to activate G-protein signaling to regulate the PC1/PC2 calcium channel.
Collapse
Affiliation(s)
- Stephen C Parnell
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Brenda S Magenheimer
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Robin L Maser
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA.,Department of Clinical Laboratory Sciences, University of Kansas Medical Center, Kansas City, KS, USA
| | - Tengis S Pavlov
- Division of Hypertension and Vascular Research, Henry Ford Hospital, Detroit, MI, USA
| | | | - Michelle L Hastings
- Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Stephen F Jackson
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Christopher J Ward
- The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kenneth R Peterson
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| | | | - James P Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA.,The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
39
|
Change in kidney volume after kidney transplantation in patients with autosomal polycystic kidney disease. PLoS One 2018; 13:e0209332. [PMID: 30589879 PMCID: PMC6307782 DOI: 10.1371/journal.pone.0209332] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 12/04/2018] [Indexed: 12/16/2022] Open
Abstract
Background The indication to bilateral nephrectomy in patients with autosomal dominant polycystic kidney scheduled for kidney transplantation is controversial. Indeed, the progressive enlargement of cysts may increase the risk of complications and the need for nephrectomy. However, very few studies investigated the change in kidney volume after kidney transplantation. Material and methods In this prospective cohort study, the change in native kidney volume in polycystic patients was evaluated with magnetic resonance imaging. Forty patients were included in the study. Kidney diameters and total kidney volume were evaluated with magnetic resonance imaging in patients who underwent simultaneous nephrectomy and kidney transplantation and in patients with kidney transplant alone, before transplantation and 1 year after transplantation. Results There was a significant reduction of kidney volume after transplantation, with a mean degree of kidney diameters reduction varying from 12.24% to 14.43%. Mean total kidney volume of the 55 kidney considered in the analysis significantly reduced from 1617.94 ± 833.42 ml to 1381.42 ± 1005.73 ml (P<0.05), with a mean rate of 16.44% of volume decrease. More than 80% of patients had a volume reduction in both groups. Conclusions Polycystic kidneys volume significantly reduces after kidney transplantation, and this would reduce the need for prophylactic bilateral nephrectomy in asymptomatic patients.
Collapse
|
40
|
Chou LF, Cheng YL, Hsieh CY, Lin CY, Yang HY, Chen YC, Hung CC, Tian YC, Yang CW, Chang MY. Effect of Trehalose Supplementation on Autophagy and Cystogenesis in a Mouse Model of Polycystic Kidney Disease. Nutrients 2018; 11:nu11010042. [PMID: 30585217 PMCID: PMC6356442 DOI: 10.3390/nu11010042] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/15/2018] [Accepted: 12/20/2018] [Indexed: 12/28/2022] Open
Abstract
Autophagy impairment has been demonstrated in the pathogenesis of autosomal dominant polycystic kidney disease (ADPKD) and could be a new target of treatment. Trehalose is a natural, nonreducing disaccharide that has been shown to enhance autophagy. Therefore, we investigated whether trehalose treatment reduces renal cyst formation in a Pkd1-hypomorphic mouse model. Pkd1 miRNA transgenic (Pkd1 miR Tg) mice and wild-type littermates were given drinking water supplemented with 2% trehalose from postnatal day 35 to postnatal day 91. The control groups received pure water or 2% sucrose for the control of hyperosmolarity. The effect on kidney weights, cystic indices, renal function, cell proliferation, and autophagic activities was determined. We found that Pkd1 miR Tg mice had a significantly lower renal mRNA expression of autophagy-related genes, including atg5, atg12, ulk1, beclin1, and p62, compared with wild-type control mice. Furthermore, immunohistochemical analysis showed that cystic lining cells had strong positive staining for the p62 protein, indicating impaired degradation of the protein by the autophagy-lysosome pathway. However, trehalose treatment did not improve reduced autophagy activities, nor did it reduce relative kidney weights, plasma blood urea nitrogen levels, or cystatin C levels in Pkd1 miR Tg mice. Histomorphological analysis revealed no significant differences in the renal cyst index, fibrosis score, or proliferative score among trehalose-, sucrose-, and water-treated groups. Our results demonstrate that adding trehalose to drinking water does not modulate autophagy activities and renal cystogenesis in Pkd1-deficient mice, suggesting that an oral supplement of trehalose may not affect the progression of ADPKD.
Collapse
Affiliation(s)
- Li-Fang Chou
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ya-Lien Cheng
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chun-Yih Hsieh
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chan-Yu Lin
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Huang-Yu Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Yung-Chang Chen
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Cheng-Chieh Hung
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ya-Chung Tian
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Chih-Wei Yang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| | - Ming-Yang Chang
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan, Taiwan.
| |
Collapse
|
41
|
Jetten AM. GLIS1-3 transcription factors: critical roles in the regulation of multiple physiological processes and diseases. Cell Mol Life Sci 2018; 75:3473-3494. [PMID: 29779043 PMCID: PMC6123274 DOI: 10.1007/s00018-018-2841-9] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022]
Abstract
Krüppel-like zinc finger proteins form one of the largest families of transcription factors. They function as key regulators of embryonic development and a wide range of other physiological processes, and are implicated in a variety of pathologies. GLI-similar 1-3 (GLIS1-3) constitute a subfamily of Krüppel-like zinc finger proteins that act either as activators or repressors of gene transcription. GLIS3 plays a critical role in the regulation of multiple biological processes and is a key regulator of pancreatic β cell generation and maturation, insulin gene expression, thyroid hormone biosynthesis, spermatogenesis, and the maintenance of normal kidney functions. Loss of GLIS3 function in humans and mice leads to the development of several pathologies, including neonatal diabetes and congenital hypothyroidism, polycystic kidney disease, and infertility. Single nucleotide polymorphisms in GLIS3 genes have been associated with increased risk of several diseases, including type 1 and type 2 diabetes, glaucoma, and neurological disorders. GLIS2 plays a critical role in the kidney and GLIS2 dysfunction leads to nephronophthisis, an end-stage, cystic renal disease. In addition, GLIS1-3 have regulatory functions in several stem/progenitor cell populations. GLIS1 and GLIS3 greatly enhance reprogramming efficiency of somatic cells into induced embryonic stem cells, while GLIS2 inhibits reprogramming. Recent studies have obtained substantial mechanistic insights into several physiological processes regulated by GLIS2 and GLIS3, while a little is still known about the physiological functions of GLIS1. The localization of some GLIS proteins to the primary cilium suggests that their activity may be regulated by a downstream primary cilium-associated signaling pathway. Insights into the upstream GLIS signaling pathway may provide opportunities for the development of new therapeutic strategies for diabetes, hypothyroidism, and other diseases.
Collapse
Affiliation(s)
- Anton M Jetten
- Cell Biology Group, Immunity, Inflammation and Disease Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC, 27709, USA.
| |
Collapse
|
42
|
Futel M, Le Bouffant R, Buisson I, Umbhauer M, Riou JF. Characterization of potential TRPP2 regulating proteins in early Xenopus embryos. J Cell Biochem 2018; 119:10338-10350. [PMID: 30171710 DOI: 10.1002/jcb.27376] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 07/02/2018] [Indexed: 11/10/2022]
Abstract
Transient receptor potential cation channel-2 (TRPP2) is a nonspecific Ca2+ -dependent cation channel with versatile functions including control of extracellular calcium entry at the plasma membrane, release of intracellular calcium ([Ca2+ ]i) from internal stores of endoplasmic reticulum, and calcium-dependent mechanosensation in the primary cilium. In early Xenopus embryos, TRPP2 is expressed in cilia of the gastrocoel roof plate (GRP) involved in the establishment of left-right asymmetry, and in nonciliated kidney field (KF) cells, where it plays a central role in early specification of nephron tubule cells dependent on [Ca2+ ]i signaling. Identification of proteins binding to TRPP2 in embryo cells can provide interesting clues about the mechanisms involved in its regulation during these various processes. Using mass spectrometry, we have therefore characterized proteins from late gastrula/early neurula stage embryos coimmunoprecipitating with TRPP2. Binding of three of these proteins, golgin A2, protein kinase-D1, and disheveled-2 has been confirmed by immunoblotting analysis of TRPP2-coprecipitated proteins. Expression analysis of the genes, respectively, encoding these proteins, golga2, prkd1, and dvl2 indicates that they are likely to play a role in these two regions. Golga2 and prkd1 are expressed at later stage in the developing pronephric tubule where golgin A2 and protein kinase-D1 might also interact with TRPP2. Colocalization experiments using exogenously expressed fluorescent versions of TRPP2 and dvl2 in GRP and KF reveal that these two proteins are generally not coexpressed, and only colocalized in discrete region of cells. This was observed in KF cells, but does not appear to occur in the apical ciliated region of GRP cells.
Collapse
Affiliation(s)
- Mélinée Futel
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, IBPS, Laboratoire de Biologie du Développement, UMR7622, 9 quai Saint-Bernard, Paris F-75005, France
| | - Ronan Le Bouffant
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, IBPS, Laboratoire de Biologie du Développement, UMR7622, 9 quai Saint-Bernard, Paris F-75005, France
| | - Isabelle Buisson
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, IBPS, Laboratoire de Biologie du Développement, UMR7622, 9 quai Saint-Bernard, Paris F-75005, France
| | - Muriel Umbhauer
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, IBPS, Laboratoire de Biologie du Développement, UMR7622, 9 quai Saint-Bernard, Paris F-75005, France
| | - Jean-François Riou
- Sorbonne Université, CNRS, Institut de Biologie Paris-Seine, IBPS, Laboratoire de Biologie du Développement, UMR7622, 9 quai Saint-Bernard, Paris F-75005, France
| |
Collapse
|
43
|
Masyuk TV, Masyuk AI, LaRusso NF. Therapeutic Targets in Polycystic Liver Disease. Curr Drug Targets 2018; 18:950-957. [PMID: 25915482 DOI: 10.2174/1389450116666150427161743] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Revised: 02/06/2015] [Accepted: 03/02/2015] [Indexed: 02/06/2023]
Abstract
Polycystic liver diseases (PLD) are a group of genetic disorders initiated by mutations in several PLD-related genes and characterized by the presence of multiple cholangiocyte-derived hepatic cysts that progressively replace liver tissue. PLD co-exists with Autosomal Dominant Polycystic Kidney Disease (ADPKD) and Autosomal Recessive PKD as well as occurs alone (i.e., Autosomal Dominant Polycystic Liver Disease [ADPLD]). PLD associated with ADPKD and ARPKD belong to a group of disorders known as cholangiociliopathies since many disease-causative and disease-related proteins are expressed in primary cilia of cholangiocytes. Aberrant expression of these proteins in primary cilia affects their structures and functions promoting cystogenesis. Current medical therapies for PLD include symptomatic management and surgical interventions. To date, the only available drug treatment for PLD patients that halt disease progression and improve quality of life are somatostatin analogs. However, the modest clinical benefits, need for long-term maintenance therapy, and the high cost of treatment justify the necessity for more effective treatment options. Substantial evidence suggests that experimental manipulations with components of the signaling pathways that influence cyst development (e.g., cAMP, intracellular calcium, receptor tyrosine kinase, transient receptor potential cation channel subfamily V member 4 (TRPV4) channel, mechanistic target of rapamycin (mTOR), histone deacetylase (HDAC6), Cdc25A phosphatase, miRNAs and metalloproteinases) attenuate growth of hepatic cysts. Many of these targets have been evaluated in pre-clinical trials suggesting their value as potential new therapies. This review outlines the current clinical and preclinical treatment strategies for PLD.
Collapse
Affiliation(s)
- Tatyana V Masyuk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Anatoliy I Masyuk
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Nicholas F LaRusso
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, 200 First Street, SW Rochester, Minnesota, MN 55905, United States
| |
Collapse
|
44
|
Shin Y, Kim DY, Ko JY, Woo YM, Park JH. Regulation of KLF12 by microRNA-20b and microRNA-106a in cystogenesis. FASEB J 2018; 32:3574-3582. [PMID: 29475398 DOI: 10.1096/fj.201700923r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common inherited disorders. ADPKD is caused by mutations in the gene encoding either polycystic kidney disease 1 ( PKD1) or polycystic kidney disease 2 ( PKD2). Patients with ADPKD show progressive growth of cystic fluid-filled renal cysts. Here, we used Pkd2f/f control mice and Pkd2f/f:HoxB7-Cre experimental mice, which are bred to have a conditional deletion of Pkd2 in the collecting ducts, and analyzed the expression pattern of microRNAs (miRNAs) of kidney tissues from Pkd2f/f and Pkd2f/f:HoxB7-Cre mice. Decreased expression of miR-20b-5p and miR-106a-5p in Pkd2f/f:HoxB7-Cre mice compared to that in Pkd2f/f mice was observed. These miRNAs target Klf12 (Krüppel-like factor 12), which has low expression in kidney tissues of Pkd2f/f mice; however, its expression is enhanced in Pkd2f/f:HoxB7-Cre mice over time. Moreover, miR-20b-5p and miR-106a-5p directly target Klf12 mRNA by binding to the 3'-UTR of Klf12. In addition, human and mouse cell lines exhibit similar patterns. These findings were also consistent with the data from Pkd2 knockout mouse embryonic fibroblasts. Furthermore, direct and indirect knockdown of Klf12 slows cyst growth and cell proliferation in mouse inner medullary collecting duct cells. Taken together, we suggest that the induction of miR-20b-5p or miR-106a-5p or the down-regulation of KLF12 could be used as potential novel therapies for inhibiting cyst growth in patients with ADPKD.-Shin, Y., Kim, D. Y., Ko, J. Y., Woo, Y. M., Park, J. H. Regulation of KLF12 by microRNA-20b and microRNA-106a in cystogenesis.
Collapse
Affiliation(s)
- Yubin Shin
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Do Yeon Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Je Yeong Ko
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Yu Mi Woo
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| | - Jong Hoon Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, South Korea
| |
Collapse
|
45
|
|
46
|
Porath B, Livingston S, Andres EL, Petrie AM, Wright JC, Woo AE, Carlton CG, Baybutt R, Vanden Heuvel GB. Cux1 promotes cell proliferation and polycystic kidney disease progression in an ADPKD mouse model. Am J Physiol Renal Physiol 2017; 313:F1050-F1059. [PMID: 28701314 PMCID: PMC5668583 DOI: 10.1152/ajprenal.00380.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 06/20/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common monogenic hereditary disorders in humans characterized by fluid-filled cysts, primarily in the kidneys. Cux1, a cell cycle regulatory gene highly expressed during kidney development, is elevated in the cyst-lining cells of Pkd1 mutant mice, and in human ADPKD cells. However, forced expression of Cux1 is insufficient to induce cystic disease in transgenic mice or to induce rapid cyst formation after cilia disruption in the kidneys of adult mice. Here we report a double mutant mouse model that has a conditional deletion of the Pkd1 gene in the renal collecting ducts together with a targeted mutation in the Cux1 gene (Pkd1CD;Cux1tm2Ejn). While kidneys isolated from newborn Pkd1CD mice exhibit cortical and medullary cysts, kidneys isolated from newborn Pkd1CD;Cux1tm2Ejn-/- mice did not show any cysts. Because Cux1tm2Ejn-/- are perinatal lethal, we evaluated Pkd1CD mice that were heterozygote for the Cux1 mutation. Similar to the newborn Pkd1CD;Cux1tm2Ejn-/- mice, newborn Pkd1CD;Cux1tm2Ejn+/- mice did not show any cysts. Comparison of Pkd1CD and Pkd1CD;Cux1tm2Ejn+/- mice at later stages of development showed a reduction in the severity of PKD in the Pkd1CD;Cux1tm2Ejn+/- mice. Moreover, we observed an increase in expression of the cyclin kinase inhibitor p27, a target of Cux1 repression, in the rescued collecting ducts. Taken together, our results suggest that Cux1 expression in PKD is not directly involved in cystogenesis but promotes cell proliferation required for expansion of existing cysts, primarily by repression of p27.
Collapse
Affiliation(s)
- Binu Porath
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | | | - Erica L Andres
- Department of Biology, Wheaton College, Wheaton, Illinois
| | | | | | - Anna E Woo
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Carol G Carlton
- Department of Anatomy and Cell Biology, University of Kansas School of Medicine, Kansas City, Kansas
| | - Richard Baybutt
- Department of Applied Health Sciences, Wheaton College, Wheaton, Illinois; and
| | - Gregory B Vanden Heuvel
- Department of Biology, Wheaton College, Wheaton, Illinois;
- Department of Biomedical Sciences, Western Michigan University Homer Stryker M.D. School of Medicine, Kalamazoo, Michigan
| |
Collapse
|
47
|
Genetic diagnosis of polycystic kidney disease, Alport syndrome, and thalassemia minor in a large Chinese family. Clin Sci (Lond) 2017; 131:2427-2438. [PMID: 28827396 DOI: 10.1042/cs20170245] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 07/31/2017] [Accepted: 08/16/2017] [Indexed: 01/13/2023]
Abstract
Polycystic kidney disease (PKD) and Alport syndrome (AS) are serious inherited disorders associated with renal disease, and thalassemia is a hereditary blood disease with a high prevalence in south China. Here, we report an exceptional PKD coincidence of thalassemia minor and AS (diagnosed genetically) in a large Chinese family. Whole genome next-generation sequencing (NGS) was performed on the proband, and all family members underwent clinical evaluation. Sanger sequencing was used to validate the mutations distinguished by NGS. The pathogenic potential of the variants were evaluated by Polymorphism Phenotyping v2 (PolyPhen-2), Sorting Intolerant From Tolerant (SIFT) algorithm, and MutationTaster. Immunohistochemical, Western blot, immunofluorescent, and TdT-mediated dUTP nick-end labeling (TUNEL) analyses were performed to investigate polycystin 1 (PC1) expression, and cell proliferation and apoptosis in kidney tissues from the proband and normal control. A novel frameshift polycystic kidney disease 1 (PKD1) mutation (c.3903delC, p.A1302Pfs) was identified to be responsible for renal disease in this family. PC1 expression, and cell proliferation and apoptosis were significantly increased in the kidney tissues of the proband. Moreover, a deletion of approximately 19.3 kb of DNA with α-globin genes (_ _SEA) was associated with thalassemia minor in the family. In addition, a collagen type IV α 5 chain (COL4A5) variant (c.2858G>T, rs78972735), annotated as a pathogenic mutation in dbSNP and human gene mutation database (HGMD), was found in four family members with no clinical traits of AS. A novel pathogenic PKD1 mutation (c.3903delC) and (_ _SEA) thalassemia deletion were found to be responsible for the clinical symptoms in this family. The reported pathogenic COL4a5 variant (c.2858G>T, rs78972735) was not pathogenic alone.
Collapse
|
48
|
Linkage Analysis of Autosomal Dominant Polycystic Kidney Disease in Iranian Families through PKD1 and PKD2 DNA Microsatellite Markers. Nephrourol Mon 2017. [DOI: 10.5812/numonthly.59996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
49
|
Ren JG, Xia HF, Yang JG, Zhu JY, Zhang W, Chen G, Zhao JH, Sun YF, Zhao YF. Down-regulation of polycystin in lymphatic malformations: possible role in the proliferation of lymphatic endothelial cells. Hum Pathol 2017; 65:231-238. [DOI: 10.1016/j.humpath.2017.05.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Revised: 04/27/2017] [Accepted: 05/10/2017] [Indexed: 02/04/2023]
|
50
|
Soroka S, Alam A, Bevilacqua M, Girard LP, Komenda P, Loertscher R, McFarlane P, Pandeya S, Tam P, Bichet DG. Assessing Risk of Disease Progression and Pharmacological Management of Autosomal Dominant Polycystic Kidney Disease: A Canadian Expert Consensus. Can J Kidney Health Dis 2017; 4:2054358117695784. [PMID: 28321325 PMCID: PMC5347414 DOI: 10.1177/2054358117695784] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited renal disorder worldwide. The disease is characterized by renal cysts and progressive renal failure due to progressive enlargement of cysts and renal fibrosis. An estimated 45% to 70% of patients with ADPKD progress to end-stage renal disease by age 65 years. Although both targeted and nontargeted therapies have been tested in patients with ADPKD, tolvaptan is currently the only pharmacological therapy approved in Canada for the treatment of ADPKD. The purpose of this consensus recommendation is to develop an evidence-informed recommendation for the optimal management of adult patients with ADPKD. This document focuses on the role of genetic testing, the role of renal imaging, predicting the risk of disease progression, and pharmacological treatment options for ADPKD. These areas of focus were derived from 2 national surveys that were disseminated to nephrologists and patients with ADPKD with the aim of identifying unmet needs in the management of ADPKD in Canada. Specific recommendations are provided for the treatment of ADPKD with tolvaptan.
Collapse
Affiliation(s)
- Steven Soroka
- Division of Nephrology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Ahsan Alam
- Division of Nephrology, Royal Victoria Hospital, McGill University, Montreal, Québec, Canada
| | - Micheli Bevilacqua
- Division of Nephrology, St. Paul’s Hospital, University of British Columbia, Vancouver, Canada
| | - Louis-Philippe Girard
- Division of Nephrology, Foothills Medical Centre, University of Calgary, Alberta, Canada
| | - Paul Komenda
- Division of Nephrology, Seven Oaks General Hospital, University of Manitoba, Winnipeg, Canada
| | - Rolf Loertscher
- Division of Nephrology, Lakeshore General Hospital, McGill University, Pointe-Claire, Québec, Canada
| | - Philip McFarlane
- Division of Nephrology, St. Michael’s Hospital, University of Toronto, Ontario, Canada
| | - Sanjaya Pandeya
- Division of Nephrology, Halton Healthcare Services, Oakville, Ontario, Canada
| | - Paul Tam
- The Scarborough Hospital, Ontario, Canada
| | - Daniel G. Bichet
- Division of Nephrology, Département de Médecine et de Physiologie Moléculaire et Intégrative, Hôpital du Sacré-Cœur de Montréal, Université de Montréal, Québec, Canada
| |
Collapse
|