1
|
Jafree DJ, Perera C, Ball M, Tolomeo D, Pomeranz G, Wilson L, Davis B, Mason WJ, Funk EM, Kolatsi-Joannou M, Polschi R, Malik S, Stewart BJ, Price KL, Mitchell H, Motallebzadeh R, Muto Y, Lees R, Needham S, Moulding D, Chandler JC, Nandanwar S, Walsh CL, Winyard PJD, Scambler PJ, Hägerling R, Clatworthy MR, Humphreys BD, Lythgoe MF, Walker-Samuel S, Woolf AS, Long DA. Microvascular aberrations found in human polycystic kidneys are an early feature in a Pkd1 mutant mouse model. Dis Model Mech 2025; 18:dmm052024. [PMID: 40114603 DOI: 10.1242/dmm.052024] [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: 07/08/2024] [Accepted: 03/13/2025] [Indexed: 03/22/2025] Open
Abstract
Therapies targeting blood vessels hold promise for autosomal dominant polycystic kidney disease (ADPKD), the most common inherited disorder causing kidney failure. However, the onset and nature of kidney vascular abnormalities in ADPKD are poorly defined. Accordingly, we employed a combination of single-cell transcriptomics; three-dimensional imaging with geometric, topological and fractal analyses; and multimodal magnetic resonance imaging with arterial spin labelling to investigate aberrant microvasculature in ADPKD kidneys. Within human ADPKD kidneys with advanced cystic pathology and excretory failure, we identified a molecularly distinct blood microvascular subpopulation, characterised by impaired angiogenic signalling and metabolic dysfunction, differing from endothelial injury profiles observed in non-cystic human kidney diseases. Next, Pkd1 mutant mouse kidneys were examined postnatally, when cystic pathology is well established, but before excretory failure. An aberrant endothelial subpopulation was also detected, concurrent with reduced cortical blood perfusion. Disorganised kidney cortical microvasculature was also present in Pkd1 mutant mouse fetal kidneys when tubular dilation begins. Thus, aberrant features of cystic kidney vasculature are harmonised between human and mouse ADPKD, supporting early targeting of the vasculature as a strategy to ameliorate ADPKD progression.
Collapse
Affiliation(s)
- Daniyal J Jafree
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
- Specialised Foundation Programme in Research, NHS East of England, Cambridge CB21 5XB, UK
| | - Charith Perera
- UCL Centre for Advanced Biomedical Imaging, University College London, London WC1E 6DD, UK
| | - Mary Ball
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Daniele Tolomeo
- UCL Centre for Advanced Biomedical Imaging, University College London, London WC1E 6DD, UK
| | - Gideon Pomeranz
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Laura Wilson
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Benjamin Davis
- Central Laser Facility, Science and Technologies Facilities Council, UK Research and Innovation, Didcot OX11 0QX, UK
| | - William J Mason
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Eva Maria Funk
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Lymphovascular Medicine and Translational 3D-Histopathology Research Group, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin 10117, Germany
| | - Maria Kolatsi-Joannou
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Radu Polschi
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Saif Malik
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Benjamin J Stewart
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Karen L Price
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Hannah Mitchell
- Mathematical Sciences Research Centre, Queen's University Belfast, Belfast BT7 1NN, UK
| | - Reza Motallebzadeh
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
- Research Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, London NW3 2PF, UK
- UCL Institute of Immunity and Transplantation, University College London, London NW3 2PF, UK
| | - Yoshiharu Muto
- Division of Nephrology, Department of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Robert Lees
- Central Laser Facility, Science and Technologies Facilities Council, UK Research and Innovation, Didcot OX11 0QX, UK
| | - Sarah Needham
- Central Laser Facility, Science and Technologies Facilities Council, UK Research and Innovation, Didcot OX11 0QX, UK
| | - Dale Moulding
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Jennie C Chandler
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Sonal Nandanwar
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Claire L Walsh
- UCL Centre for Advanced Biomedical Imaging, University College London, London WC1E 6DD, UK
- Department of Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - Paul J D Winyard
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| | - Peter J Scambler
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - René Hägerling
- Lymphovascular Medicine and Translational 3D-Histopathology Research Group, Charité Universitätsmedizin Berlin, Berlin 10117, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin 10117, Germany
| | - Menna R Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge CB2 1TN, UK
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton CB10 1SA, UK
| | - Benjamin D Humphreys
- Division of Nephrology, Department of Medicine, Washington University in St Louis, St Louis, MO 63110, USA
| | - Mark F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, University College London, London WC1E 6DD, UK
| | - Simon Walker-Samuel
- UCL Centre for Advanced Biomedical Imaging, University College London, London WC1E 6DD, UK
| | - Adrian S Woolf
- School of Biological Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - David A Long
- Developmental Biology and Cancer Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- UCL Centre for Kidney and Bladder Health, University College London, London WC1E 6BT, UK
| |
Collapse
|
2
|
Fan Q, Hadla M, Peterson Z, Nelson G, Ye H, Wang X, Mardirossian JM, Harris PC, Alper SL, Prakash YS, Beyder A, Torres VE, Chebib FT. Activation of PIEZO1 Attenuates Kidney Cystogenesis In Vitro and Ex Vivo. KIDNEY360 2024; 5:1601-1612. [PMID: 39356563 DOI: 10.34067/kid.0000000598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 09/24/2024] [Indexed: 10/04/2024]
Abstract
Key Points
PIEZO1 activation reduces cystogenesis: Yoda1 activates PIEZO1, raising calcium and lowering cAMP, reducing cyst growth in autosomal dominant polycystic kidney disease models.Context-dependent role of PIEZO1: PIEZO1 knockout in mice with or without polycystic kidneys does not affect cyst formation, suggesting redundancy in mechanosensitive pathways.Therapeutic potential: Findings support PIEZO1 activation as a part of combination therapy to slow cyst growth in autosomal dominant polycystic kidney disease, needing more in vivo studies.
Background
The disruption of calcium signaling associated with polycystin deficiency is a key factor in abnormal epithelial growth in autosomal dominant polycystic kidney disease. Calcium homeostasis can be influenced by mechanotransduction. The mechanosensitive cation channel PIEZO1 has been implicated in sensing intrarenal pressure and regulating urinary osmoregulation, but its role in kidney cystogenesis is unclear.
Methods
We hypothesized that altered mechanotransduction contributes to cystogenesis in autosomal dominant polycystic kidney disease and that activation of mechanosensitive cation channels could be a therapeutic strategy.
Results
We demonstrate that Yoda1, a PIEZO1 activator, increases intracellular calcium and reduces forskolin-induced cAMP levels in mouse inner medullary collecting duct (mIMCD3) cells. Notably, knockout of polycystin-2 attenuated the efficacy of Yoda1 in reducing cAMP levels in mIMCD3 cells. Yoda1 also reduced forskolin-induced mIMCD3 cyst surface area in vitro and cystic index in mouse metanephros ex vivo in a dose-dependent manner. However, collecting duct–specific PIEZO1 knockout neither induced cystogenesis in wild-type mice nor altered cystogenesis in the Pkd1RC/RC mouse model.
Conclusions
These findings support the potential role of PIEZO1 agonists in mitigating cystogenesis by increasing intracellular calcium and reducing cAMP levels, but the unaltered in vivo cystic phenotype after PIEZO1 knockout in the collecting duct suggests possible redundancy in mechanotransductive pathways.
Collapse
Affiliation(s)
- Qingfeng Fan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Mohamad Hadla
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | - Zack Peterson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Grace Nelson
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Hong Ye
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular biology, Mayo Clinic, Rochester, Minnesota
| | - Seth L Alper
- Division of Nephrology and Vascular Biology Research Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Y S Prakash
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
| | - Arthur Beyder
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota
- Enteric NeuroScience Program (ENSP), Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Fouad T Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
3
|
Fan Q, Hadla M, Peterson Z, Nelson G, Ye H, Wang X, Mardirossian JM, Harris PC, Alper SL, Prakash YS, Beyder A, Torres VE, Chebib FT. Activation of Piezo1 Inhibits Kidney Cystogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593717. [PMID: 38766249 PMCID: PMC11101129 DOI: 10.1101/2024.05.11.593717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
The disruption of calcium signaling associated with polycystin deficiency has been proposed as the primary event underlying the increased abnormally patterned epithelial cell growth characteristic of Polycystic Kidney Disease. Calcium can be regulated through mechanotransduction, and the mechanosensitive cation channel Piezo1 has been implicated in sensing of intrarenal pressure and in urinary osmoregulation. However, a possible role for PIEZO1 in kidney cystogenesis remains undefined. We hypothesized that cystogenesis in ADPKD reflects altered mechanotransduction, suggesting activation of mechanosensitive cation channels as a therapeutic strategy for ADPKD. Here, we show that Yoda-1 activation of PIEZO1 increases intracellular Ca 2+ and reduces forskolin-induced cAMP levels in mIMCD3 cells. Yoda-1 reduced forskolin-induced IMCD cyst surface area in vitro and in mouse metanephros ex vivo in a dose-dependent manner. Knockout of polycystin-2 dampened the efficacy of PIEZO1 activation in reducing both cAMP levels and cyst surface area in IMCD3 cells. However, collecting duct-specific Piezo1 knockout neither induced cystogenesis in wild-type mice nor affected cystogenesis in the Pkd1 RC/RC model of ADPKD. Our study suggests that polycystin-2 and PIEZO1 play a role in mechanotransduction during cystogenesis in vitro , and ex vivo , but that in vivo cyst expansion may require inactivation or repression of additional suppressors of cystogenesis and/or growth. Our study provides a preliminary proof of concept for PIEZO1 activation as a possible component of combination chemotherapy to retard or halt cystogenesis and/or cyst growth.
Collapse
|
4
|
Mbiakop UC, Jaggar JH. Vascular polycystin proteins in health and disease. Microcirculation 2024; 31:e12834. [PMID: 37823335 PMCID: PMC11009377 DOI: 10.1111/micc.12834] [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: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
PKD1 (polycystin 1) and PKD2 (polycystin 2) are expressed in a variety of different cell types, including arterial smooth muscle and endothelial cells. PKD1 is a transmembrane domain protein with a large extracellular N-terminus that is proposed to act as a mechanosensor and receptor. PKD2 is a member of the transient receptor potential (TRP) channel superfamily which is also termed TRPP1. Mutations in the genes which encode PKD1 and PKD2 lead to autosomal dominant polycystic kidney disease (ADPKD). ADPKD is one of the most prevalent monogenic disorders in humans and is associated with extrarenal and vascular complications, including hypertension. Recent studies have uncovered mechanisms of activation and physiological functions of PKD1 and PKD2 in arterial smooth muscle and endothelial cells. It has also been found that PKD function is altered in the vasculature during ADPKD and hypertension. We will summarize this work and discuss future possibilities for this area of research.
Collapse
Affiliation(s)
- Ulrich C. Mbiakop
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38163
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38163
| |
Collapse
|
5
|
Righini M, Mancini R, Busutti M, Buscaroli A. Autosomal Dominant Polycystic Kidney Disease: Extrarenal Involvement. Int J Mol Sci 2024; 25:2554. [PMID: 38473800 DOI: 10.3390/ijms25052554] [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: 01/03/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disorder, but kidneys are not the only organs involved in this systemic disorder. Individuals with the condition may display additional manifestations beyond the renal system, involving the liver, pancreas, and brain in the context of cystic manifestations, while involving the vascular system, gastrointestinal tract, bones, and cardiac valves in the context of non-cystic manifestations. Despite kidney involvement remaining the main feature of the disease, thanks to longer survival, early diagnosis, and better management of kidney-related problems, a new wave of complications must be faced by clinicians who treated patients with ADPKD. Involvement of the liver represents the most prevalent extrarenal manifestation and has growing importance in the symptom burden and quality of life. Vascular abnormalities are a key factor for patients' life expectancy and there is still debate whether to screen or not to screen all patients. Arterial hypertension is often the earliest onset symptom among ADPKD patients, leading to frequent cardiovascular complications. Although cardiac valvular abnormalities are a frequent complication, they rarely lead to relevant problems in the clinical history of polycystic patients. One of the newest relevant aspects concerns bone disorders that can exert a considerable influence on the clinical course of these patients. This review aims to provide the "state of the art" among the extrarenal manifestation of ADPKD.
Collapse
Affiliation(s)
- Matteo Righini
- Nephrology and Dialysis Unit, Santa Maria delle Croci Hospital, AUSL Romagna, 48121 Ravenna, Italy
- Nephrology, Dialysis and Transplantation Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Raul Mancini
- Nephrology, Dialysis and Transplantation Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Marco Busutti
- Nephrology, Dialysis and Transplantation Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Andrea Buscaroli
- Nephrology and Dialysis Unit, Santa Maria delle Croci Hospital, AUSL Romagna, 48121 Ravenna, Italy
| |
Collapse
|
6
|
Clearman KR, Haycraft CJ, Croyle MJ, Collawn JF, Yoder BK. Functions of the primary cilium in the kidney and its connection with renal diseases. Curr Top Dev Biol 2023; 155:39-94. [PMID: 38043952 DOI: 10.1016/bs.ctdb.2023.07.001] [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] [Indexed: 12/05/2023]
Abstract
The nonmotile primary cilium is a sensory structure found on most mammalian cell types that integrates multiple signaling pathways involved in tissue development and postnatal function. As such, mutations disrupting cilia activities cause a group of disorders referred to as ciliopathies. These disorders exhibit a wide spectrum of phenotypes impacting nearly every tissue. In the kidney, primary cilia dysfunction caused by mutations in polycystin 1 (Pkd1), polycystin 2 (Pkd2), or polycystic kidney and hepatic disease 1 (Pkhd1), result in polycystic kidney disease (PKD), a progressive disorder causing renal functional decline and end-stage renal disease. PKD affects nearly 1 in 1000 individuals and as there is no cure for PKD, patients frequently require dialysis or renal transplantation. Pkd1, Pkd2, and Pkhd1 encode membrane proteins that all localize in the cilium. Pkd1 and Pkd2 function as a nonselective cation channel complex while Pkhd1 protein function remains uncertain. Data indicate that the cilium may act as a mechanosensor to detect fluid movement through renal tubules. Other functions proposed for the cilium and PKD proteins in cyst development involve regulation of cell cycle and oriented division, regulation of renal inflammation and repair processes, maintenance of epithelial cell differentiation, and regulation of mitochondrial structure and metabolism. However, how loss of cilia or cilia function leads to cyst development remains elusive. Studies directed at understanding the roles of Pkd1, Pkd2, and Pkhd1 in the cilium and other locations within the cell will be important for developing therapeutic strategies to slow cyst progression.
Collapse
Affiliation(s)
- Kelsey R Clearman
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Courtney J Haycraft
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Mandy J Croyle
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James F Collawn
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bradley K Yoder
- Department of Cell, Developmental, and Integrative Biology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
7
|
Lin CC, Menezes LF, Qiu J, Pearson E, Zhou F, Ishimoto Y, Anderson DE, Germino GG. In vivo Polycystin-1 interactome using a novel Pkd1 knock-in mouse model. PLoS One 2023; 18:e0289778. [PMID: 37540694 PMCID: PMC10403143 DOI: 10.1371/journal.pone.0289778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023] Open
Abstract
PKD1 is the most commonly mutated gene causing autosomal dominant polycystic kidney disease (ADPKD). It encodes Polycystin-1 (PC1), a putative membrane protein that undergoes a set of incompletely characterized post-transcriptional cleavage steps and has been reported to localize in multiple subcellular locations, including the primary cilium and mitochondria. However, direct visualization of PC1 and detailed characterization of its binding partners remain challenging. We now report a new mouse model with HA epitopes and eGFP knocked-in frame into the endogenous mouse Pkd1 gene by CRISPR/Cas9. Using this model, we sought to visualize endogenous PC1-eGFP and performed affinity-purification mass spectrometry (AP-MS) and network analyses. We show that the modified Pkd1 allele is fully functional but the eGFP-tagged protein cannot be detected without signal amplification by secondary antibodies. Using nanobody-coupled beads and large quantities of tissue, AP-MS identified an in vivo PC1 interactome, which is enriched for mitochondrial proteins and components of metabolic pathways. These studies suggest this mouse model and interactome data will be useful to understand PC1 function, but that new methods and brighter tags will be required to track endogenous PC1.
Collapse
Affiliation(s)
- Cheng-Chao Lin
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Luis F. Menezes
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Jiahe Qiu
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Elisabeth Pearson
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Fang Zhou
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yu Ishimoto
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - D. Eric Anderson
- Advanced Mass Spectrometry Core, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Gregory G. Germino
- Polycystic Kidney Disease Section, Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
8
|
Kurbegovic A, Pacis RC, Trudel M. Modeling Pkd1 gene-targeted strategies for correction of polycystic kidney disease. Mol Ther Methods Clin Dev 2023; 29:366-380. [PMID: 37214311 PMCID: PMC10199405 DOI: 10.1016/j.omtm.2023.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 03/31/2023] [Indexed: 04/05/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) causes renal cysts and leads to end-stage renal disease in midlife due mainly to PKD1 gene mutations. Virtually no studies have explored gene therapeutic strategies for long-term effective treatment of PKD. Toward this aim, the severely cystic Pkd1-null mouse model was targeted with a series of transgene transfers using genomic Pkd1 under its regulatory elements (Pkd1wt), a kidney-targeted Pkd1 gene (SBPkd1), or Pkd1Minigene. The introduced Pkd1wt gene constructs with ∼8-fold overexpression display similar endogenous cellular profiles and full complementation of Pkd1-/- phenotype and establish the referral Pkd1 genomic length for proper regulation. SBPkd1 transgene transfer expressing 0.6- or 7-fold Pkd1 endogenous levels is sufficient to correct glomerular and proximal tubular cysts and to markedly postpone cysts in other tubular segments as well, showing that the small SB elements appreciably overlap with Pkd1 promoter/5' UTR regulation. Renal-targeted Pkd1Minigene at high copy numbers conveys an expression level similar to that of the endogenous Pkd1 gene, with widespread and homogeneous weak Pkd1 cellular signal, partially rescuing all cystic tubular segments. These transgene transfers determine that Pkd1 intragenic sequences regulate not only expression levels but also spatiotemporal patterns. Importantly, our study demonstrates that Pkd1 re-expression from hybrid therapeutic constructs can ameliorate, with considerably extended lifespan, or eliminate PKD.
Collapse
Affiliation(s)
- Almira Kurbegovic
- Institut de Recherches Cliniques de Montréal, Faculté de, Médecine, Montreal, QC, Canada
| | - Rey Christian Pacis
- Institut de Recherches Cliniques de Montréal, Faculté de, Médecine, Montreal, QC, Canada
| | - Marie Trudel
- Institut de Recherches Cliniques de Montréal, Faculté de, Médecine, Montreal, QC, Canada
| |
Collapse
|
9
|
Qiu J, Germino GG, Menezes LF. Mechanisms of Cyst Development in Polycystic Kidney Disease. ADVANCES IN KIDNEY DISEASE AND HEALTH 2023; 30:209-219. [PMID: 37088523 PMCID: PMC10289784 DOI: 10.1053/j.akdh.2023.03.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Autosomal dominant polycystic kidney disease is the most common inherited cause of end-stage kidney disease worldwide. Most cases result from mutation of either of 2 genes, PKD1 and PKD2, which encode proteins that form a probable receptor/channel complex. Studies suggest that a loss of function of the complex below an indeterminate threshold triggers cyst initiation, which ultimately results in dysregulation of multiple metabolic processes and downstream pathways and subsequent cyst growth. Noncell autonomous factors may also promote cyst growth. In this report, we focus primarily on the process of early cyst formation and factors that contribute to its variability with brief consideration of how new studies suggest this process may be reversible.
Collapse
Affiliation(s)
- Jiahe Qiu
- Polycystic Kidney Disease Section, Kidney Disease Branch, Division of Intramural Research, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Gregory G Germino
- Polycystic Kidney Disease Section, Kidney Disease Branch, Division of Intramural Research, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD.
| | - Luis F Menezes
- Polycystic Kidney Disease Section, Kidney Disease Branch, Division of Intramural Research, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD.
| |
Collapse
|
10
|
Papavassiliou KA, Gargalionis AN, Papavassiliou AG. Polycystins, mechanotransduction and cancer development. J Cell Mol Med 2022; 26:2741-2743. [PMID: 35366054 PMCID: PMC9077297 DOI: 10.1111/jcmm.17298] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/23/2022] [Indexed: 12/11/2022] Open
Affiliation(s)
- Kostas A. Papavassiliou
- Department of Biological ChemistryMedical SchoolNational and Kapodistrian University of AthensAthensGreece
| | - Antonios N. Gargalionis
- Department of Biological ChemistryMedical SchoolNational and Kapodistrian University of AthensAthensGreece
| | | |
Collapse
|
11
|
Lee WJ, Jung KH, Ryu H, Oh KH, Kim JM, Lee ST, Park KI, Chu K, Jung KY, Kim M, Lee SK. Association of autosomal dominant polycystic kidney disease with cerebral small vessel disease. J Cereb Blood Flow Metab 2021; 41:3365-3377. [PMID: 34415212 PMCID: PMC8669289 DOI: 10.1177/0271678x211037869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Cilia dysfunction in autosomal-dominant polycystic kidney disease (ADPKD) may impair the integrity of glymphatic system and be implicated in the progression of cerebral small vessel disease (SVD), although the link between the two diseases has not been investigated. We evaluated the association of ADPKD pathology with SVD pattern and severity. Overall, 304 individuals in an ADPKD (chronic kidney disease stage ≤4 and age ≥50 years) cohort and their age, sex, and estimated glomerular filtration rate (eGFR)-matched controls were retrospectively included. ADPKD severity was classified into 1 A-B, 1 C, and 1 D-E, according to age and height-adjusted total kidney volume. SVD parameters included white-matter hyperintensity (WMH) severity scale, enlarged perivascular space (ePVS) score, and degree of lacunes or cerebral microbleeds (CMBs). After adjustments for age, sex, eGFR, and cerebrovascular risk factor parameters, ADPKD was associated with higher ePVS scores (P < 0.001), but not with the WMH severity or degree of lacunes or CMBs. In the ADPKD subgroup, higher ADPKD severity class was associated with higher ePVS scores (P < 0.001), WMH severity (P = 0.003), and degree of lacunes (P = 0.002). ADPKD associated cilia dysfunction may induce chronic cerebral glymphatic system dysfunction, which may contribute to the specific progression of ePVS compared with other SVD markers.
Collapse
Affiliation(s)
- Woo-Jin Lee
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Keun-Hwa Jung
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Hyunjin Ryu
- Department of Neurology, Seoul National University College of Medicine, Seoul, South Korea
| | - Kook-Hwan Oh
- Department of Neurology, Seoul National University College of Medicine, Seoul, South Korea
| | - Jeong-Min Kim
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Soon-Tae Lee
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Kyung-Il Park
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea.,Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Kon Chu
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Ki-Young Jung
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| | - Manho Kim
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea.,Department of Neurology, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, South Korea
| | - Sang Kun Lee
- Department of Neurology, Seoul National University Hospital, Seoul, South Korea
| |
Collapse
|
12
|
Carullo N, Zicarelli MT, Casarella A, Nicotera R, Castagna A, Urso A, Presta P, Andreucci M, Russo E, Bolignano D, Coppolino G. Retarding Progression of Chronic Kidney Disease in Autosomal Dominant Polycystic Kidney Disease with Metformin and Other Therapies: An Update of New Insights. Int J Gen Med 2021; 14:5993-6000. [PMID: 34588803 PMCID: PMC8473846 DOI: 10.2147/ijgm.s305491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/13/2021] [Indexed: 12/13/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent single-gene disorder leading to renal failure. Current therapies are aimed to treat renal and extrarenal complications of ADPKD, but improved knowledge of the pathophysiological mechanisms leading to the generation and growth of cysts has permitted the identification of new drug candidates for clinical trials. Among these, in this review, we will examine above all the role of metformin, hypothesized to be able to activate the AMP-activated protein kinase (AMPK) pathway and potentially modulate some mechanisms implicated in the onset and the growth of the cysts.
Collapse
Affiliation(s)
- Nazareno Carullo
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | | | | | - Ramona Nicotera
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Alberto Castagna
- Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Alessandra Urso
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Pierangela Presta
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Michele Andreucci
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Emilio Russo
- Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Davide Bolignano
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| | - Giuseppe Coppolino
- Renal Unit, Department of Health Sciences, "Magna Graecia" University, Catanzaro, Italy
| |
Collapse
|
13
|
The cellular pathways and potential therapeutics of Polycystic Kidney Disease. Biochem Soc Trans 2021; 49:1171-1188. [PMID: 34156429 DOI: 10.1042/bst20200757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 02/07/2023]
Abstract
Polycystic Kidney Disease (PKD) refers to a group of disorders, driven by the formation of cysts in renal tubular cells and is currently one of the leading causes of end-stage renal disease. The range of symptoms observed in PKD is due to mutations in cilia-localising genes, resulting in changes in cellular signalling. As such, compounds that are currently in preclinical and clinical trials target some of these signalling pathways that are dysregulated in PKD. In this review, we highlight these pathways including cAMP, EGF and AMPK signalling and drugs that target them and may show promise in lessening the disease burden of PKD patients. At present, tolvaptan is the only approved therapy for ADPKD, however, it carries several adverse side effects whilst comparatively, no pharmacological drug is approved for ARPKD treatment. Aside from this, drugs that have been the subject of multiple clinical trials such as metformin, which targets AMPK signalling and somatostatins, which target cAMP signalling have shown great promise in reducing cyst formation and cellular proliferation. This review also discusses other potential and novel targets that can be used for future interventions, such as β-catenin and TAZ, where research has shown that a reduction in the overexpression of these signalling components results in amelioration of disease phenotype. Thus, it becomes apparent that well-designed preclinical investigations and future clinical trials into these pathways and other potential signalling targets are crucial in bettering disease prognosis for PKD patients and could lead to personalised therapy approaches.
Collapse
|
14
|
Cardiac Involvement in Autosomal Dominant Polycystic Kidney Disease. CARDIOGENETICS 2021. [DOI: 10.3390/cardiogenetics11020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Cardiovascular disorders are the main complication in autosomal dominant polycystic kidney disease (ADPKD). contributing to both morbidity and mortality. This review considers clinical studies unveiling cardiovascular features in patients with ADPKD. Additionally, it focuses on basic science studies addressing the dysfunction of the polycystin proteins located in the cardiovascular system as a contributing factor to cardiovascular abnormalities. In particular, the effects of polycystin proteins’ deficiency on the cardiomyocyte function have been considered.
Collapse
|
15
|
Nigro EA, Boletta A. Role of the polycystins as mechanosensors of extracellular stiffness. Am J Physiol Renal Physiol 2021; 320:F693-F705. [PMID: 33615892 DOI: 10.1152/ajprenal.00545.2020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Polycystin-1 (PC-1) is a transmembrane protein, encoded by the PKD1 gene, mutated in autosomal dominant polycystic kidney disease (ADPKD). This common genetic disorder, characterized by cyst formation in both kidneys, ultimately leading to renal failure, is still waiting for a definitive treatment. The overall function of PC-1 and the molecular mechanism responsible for cyst formation are slowly coming to light, but they are both still intensively studied. In particular, PC-1 has been proposed to act as a mechanosensor, although the precise signal that activates the mechanical properties of this protein has been long debated and questioned. In this review, we report studies and evidence of PC-1 function as a mechanosensor, starting from the peculiarity of its structure, through the long journey that progressively shed new light on the potential initiating events of cystogenesis, concluding with the description of PC-1 recently shown ability to sense the mechanical stimuli provided by the stiffness of the extracellular environment. These new findings have potentially important implications for the understanding of ADPKD pathophysiology and potentially for designing new therapies.NEW & NOTEWORTHY Polycystin-1 has recently emerged as a possible receptor able to sense extracellular stiffness and to negatively control the cellular actomyosin contraction machinery. Here, we revisit a large body of literature on autosomal dominant polycystic kidney disease providing a new possible mechanistic view on the topic.
Collapse
Affiliation(s)
- Elisa A Nigro
- Molecular Basis of Cystic Kidney Diseases, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Boletta
- Molecular Basis of Cystic Kidney Diseases, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
16
|
Hu J, Harris PC. Regulation of polycystin expression, maturation and trafficking. Cell Signal 2020; 72:109630. [PMID: 32275942 PMCID: PMC7269868 DOI: 10.1016/j.cellsig.2020.109630] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/26/2022]
Abstract
The major autosomal dominant polycystic kidney disease (ADPKD) genes, PKD1 and PKD2, are wildly expressed at the organ and tissue level. PKD1 encodes polycystin 1 (PC1), a large membrane associated receptor-like protein that can complex with the PKD2 product, PC2. Various cellular locations have been described for both PC1, including the plasma membrane and extracellular vesicles, and PC2, especially the endoplasmic reticulum (ER), but compelling evidence indicates that the primary cilium, a sensory organelle, is the key site for the polycystin complex to prevent PKD. As with other membrane proteins, the ER biogenesis pathway is key to appropriately folding, performing quality control, and exporting fully folded PC1 to the Golgi apparatus. There is a requirement for binding with PC2 and cleavage of PC1 at the GPS for this folding and export to occur. Six different monogenic defects in this pathway lead to cystic disease development, with PC1 apparently particularly sensitive to defects in this general protein processing pathway. Trafficking of membrane proteins, and the polycystins in particular, through the Golgi to the primary cilium have been analyzed in detail, but at this time, there is no clear consensus on a ciliary targeting sequence required to export proteins to the cilium. After transitioning though the trans-Golgi network, polycystin-bearing vesicles are likely sorted to early or recycling endosomes and then transported to the ciliary base, possibly via docking to transition fibers (TF). The membrane-bound polycystin complex then undergoes facilitated trafficking through the transition zone, the diffusion barrier at the base of the cilium, before entering the cilium. Intraflagellar transport (IFT) may be involved in moving the polycystins along the cilia, but data also indicates other mechanisms. The ciliary polycystin complex can be ubiquitinated and removed from cilia by internalization at the ciliary base and may be sent back to the plasma membrane for recycling or to lysosomes for degradation. Monogenic defects in processes regulating the protein composition of cilia are associated with syndromic disorders involving many organ systems, reflecting the pleotropic role of cilia during development and for tissue maintenance. Many of these ciliopathies have renal involvement, likely because of faulty polycystin signaling from cilia. Understanding the expression, maturation and trafficking of the polycystins helps understand PKD pathogenesis and suggests opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Jinghua Hu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| | - Peter C Harris
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA; Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
17
|
Padovano V, Mistry K, Merrick D, Gresko N, Caplan MJ. A cut above (and below): Protein cleavage in the regulation of polycystin trafficking and signaling. Cell Signal 2020; 72:109634. [PMID: 32283256 PMCID: PMC7269866 DOI: 10.1016/j.cellsig.2020.109634] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 10/24/2022]
Abstract
The polycystin-1 and 2 proteins, encoded by the genes mutated in Autosomal Dominant Polycystic Kidney Disease, are connected to a large number of biological pathways. While the nature of these connections and their relevance to the primary functions of the polycystin proteins have yet to be fully elucidated, it is clear that many of them are mediated by or depend upon cleavage of the polycystin-1 protein. Cleavage of polycystin-1 at its G protein coupled receptor proteolytic site is an obligate step in the protein's maturation and in aspects of its trafficking. This cleavage may also serve to prime polycystin-1 to play a role as a non-canonical G protein coupled receptor. Cleavage of the cytoplasmic polycystin-1C terminal tail releases fragments that are able to enter the nucleus and the mitochondria and to influence their activities. Understanding the nature of these cleavages, their regulation and their consequences is likely to provide valuable insights into both the physiological functions served by the polycystin proteins and the pathological consequences of their absence.
Collapse
Affiliation(s)
- Valeria Padovano
- Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, 415 Main Street, Cambridge, MA 02142, USA
| | - Kavita Mistry
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026, USA
| | - David Merrick
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026, USA
| | - Nikolay Gresko
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026, USA
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520-8026, USA.
| |
Collapse
|
18
|
Parrot C, Kurbegovic A, Yao G, Couillard M, Côté O, Trudel M. c-Myc is a regulator of the PKD1 gene and PC1-induced pathogenesis. Hum Mol Genet 2020; 28:751-763. [PMID: 30388220 DOI: 10.1093/hmg/ddy379] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/28/2018] [Accepted: 10/26/2018] [Indexed: 12/12/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is among the most common monogenic disorders mainly associated with PKD1/PC1 mutations. We show herein that renal regulation in Pc1 dosage-reduced and -increased mouse models converge toward stimulation of c-Myc expression along with β-catenin, delineating c-Myc as a key Pkd1 node in cystogenesis. Enhanced renal c-Myc-induced ADPKD in SBM transgenic mice lead conversely to striking upregulation of Pkd1/Pc1 expression and β-catenin activation, lending credence for reciprocal crosstalk between c-Myc and Pc1. In adult SBM kidneys, c-Myc is strongly enriched on Pkd1 promoter with RNA pol II, consistent with Pkd1 upregulation during cystogenesis. Similar c-Myc direct binding at birth uncovers an equivalent role on Pkd1 regulation during renal developmental program. Concurrent with enriched c-Myc binding, recruitment of active chromatin modifying co-factors by c-Myc at the Pkd1 regulatory region probably opens chromatin to stimulate transcription. A similar transcriptional activation by c-Myc is also likely operant on endogenous human PKD1 gene from our transactivation analysis in response to human c-MYC upregulation. Genetic ablation of c-Myc in Pc1-reduced and -increased mouse models significantly attenuates cyst growth, proliferation and PKD progression. Our study determined a dual role for c-Myc, as a major contributor in Pc1-induced cystogenesis and in a feed-forward regulatory Pkd1-c-Myc loop mechanism that may also prevail in human ADPKD.
Collapse
Affiliation(s)
- Camila Parrot
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Almira Kurbegovic
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Guanhan Yao
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Martin Couillard
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Olivier Côté
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| | - Marie Trudel
- Institut de recherches cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine de L'Université de Montréal, Montreal, Québec, Canada
| |
Collapse
|
19
|
Kurbegovic A, Trudel M. The master regulators Myc and p53 cellular signaling and functions in polycystic kidney disease. Cell Signal 2020; 71:109594. [PMID: 32145315 DOI: 10.1016/j.cellsig.2020.109594] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 03/02/2020] [Accepted: 03/03/2020] [Indexed: 01/08/2023]
Abstract
The transcription factors Myc and p53 associated with oncogenesis play determinant roles in a human genetic disorder, autosomal dominant polycystic kidney disease (ADPKD), that was coined early in ADPKD etiology a «neoplasia in disguise ». These factors are interdependent master cell regulators of major biological processes including proliferation, apoptosis, cell growth, metabolism, inflammation, fibrosis and differentiation that are all modulated in ADPKD. Myc and p53 proteins evolved to respond and carry out overlapping functions via opposing mechanisms of action. Studies in human ADPKD kidneys, caused by mutations in the PKD1 or PKD2 genes, reveal reduced p53 expression and high expression of Myc in the cystic tubular epithelium. Myc and p53 via direct interaction act respectively, as transcriptional activator and repressor of PKD1 gene expression, consistent with increased renal PKD1 levels in ADPKD. Mouse models generated by Pkd1 and Pkd2 gene dosage dysregulation reproduce renal cystogenesis with activation of Myc expression and numerous signaling pathways, strikingly similar to those determined in human ADPKD. In fact, upregulation of renal Myc expression is also detected in virtually all non-orthologous animal models of PKD. A definitive causal connection of Myc with cystogenesis was established by renal overexpression of Myc in transgenic mice that phenocopies human ADPKD. The network of activated signaling pathways in human and mouse cystogenesis individually or in combination can target Myc as a central node of PKD pathogenesis. One or many of the multiple functions of Myc upon activation can play a role in every phases of ADPKD development and lend credence to the notion of "Myc addiction" for cystogenesis. We propose that the residual p53 levels are conducive to an ADPKD biological program without cancerogenesis while a "p53 dependent annihilation" mechanism would be permissive to oncogenesis. Of major importance, Myc ablation in orthologous mouse models or direct inhibition in non-orthologous mouse model significantly delays cystogenesis consistent with pharmacologic or genetic inhibition of Myc upstream regulator or downstream targets in the mouse. Together, these studies on PKD proteins upon dysregulation not only converged on Myc as a focal point but also attribute to Myc upregulation a causal and « driver » role in pathogenesis. This review will present and discuss our current knowledge on Myc and p53, focused on PKD mouse models and ADPKD.
Collapse
Affiliation(s)
- Almira Kurbegovic
- Institut de Recherches Cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada
| | - Marie Trudel
- Institut de Recherches Cliniques de Montréal, Molecular Genetics and Development, Faculté de Médecine, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
20
|
Metabolism and mitochondria in polycystic kidney disease research and therapy. Nat Rev Nephrol 2019; 14:678-687. [PMID: 30120380 DOI: 10.1038/s41581-018-0051-1] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common, potentially lethal, monogenic diseases and is caused predominantly by mutations in polycystic kidney disease 1 (PKD1) and PKD2, which encode polycystin 1 (PC1) and PC2, respectively. Over the decades-long course of the disease, patients develop large fluid-filled renal cysts that impair kidney function, leading to end-stage renal disease in ~50% of patients. Despite the identification of numerous dysregulated pathways in ADPKD, the molecular mechanisms underlying the renal dysfunction from mutations in PKD genes and the physiological functions of the polycystin proteins are still unclear. Alterations in cell metabolism have emerged in the past decade as a hallmark of ADPKD. ADPKD cells shift their mode of energy production from oxidative phosphorylation to alternative pathways, such as glycolysis. In addition, the polycystins seem to play regulatory roles in modulating mechanisms and machinery related to energy production and utilization, including AMPK, PPARα, PGC1α, calcium signalling at mitochondria-associated membranes, mTORC1, cAMP and CFTR-mediated ion transport as well as the expression of crucial components of the mitochondrial energy production apparatus. In this Review, we explore these metabolic changes and discuss in detail the relationship between energy metabolism and ADPKD pathogenesis and identify potential therapeutic targets.
Collapse
|
21
|
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
|
22
|
Abstract
In Strongylocentrotus purpuratus, the fucose sulphate polymer (FSP) of egg jelly induces the sperm acrosome reaction (AR; Vacquier & Moy, 1997). Protease treatment of sperm renders the cells insensitive to FSP, indicating that sperm membrane receptors mediate the signal transduction events underlying the AR. Monoclonal antibodies to a 210 kDa membrane glycoprotein induce Ca2+ influx into sperm and trigger the AR (Trimmer et al., 1986; Moy et al., 1996). Purified 210 kDa protein binds species-specifically to egg jelly and blocks AR induction by antibody (Podell & Vacquier, 1985; Moy et al., 1996). FSP binds to the 210 kDa protein attached to Sepharose (Vacquier & Moy, 1997). Monoclonal antibodies localise the 210 kDa protein on the plasma membrane over the acrosome and also on the sperm flagellum. The 210 kDa protein has the attributes of a sperm receptor for egg jelly and is henceforth named suREJ1 (Moy et al., 1996). We describe here the three REJ proteins found thus far in S. purpuratus sperm.
Collapse
|
23
|
A case of congenital left ventricular diverticulum in a patient with autosomal dominant polycystic kidney disease: possible mechanistic link between polycystin and ventricular diverticulum. CEN Case Rep 2018; 7:237-242. [PMID: 29876750 DOI: 10.1007/s13730-018-0335-0] [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: 02/11/2018] [Accepted: 04/30/2018] [Indexed: 10/14/2022] Open
Abstract
A 40-year-old woman had been followed as an outpatient to manage chronic kidney disease secondary to autosomal dominant polycystic kidney disease (ADPKD). Atrial premature contraction was found incidentally on an electrocardiogram during her regular follow-up examination. Subsequent transthoracic echocardiography detected an abnormal structure located very close to the left ventricular outflow tract (23 mm long × 15 mm wide in diastole). The structure was finally diagnosed as congenital left ventricular diverticulum (CLVD) using transesophageal echocardiography, contrast-enhanced computed tomography, and magnetic resonance imaging. Although CLVD occasionally causes intraventricular coagulation, lethal arrhythmia, and congestive heart failure, the size and location of her diverticulum remained unchanged over time and a 24-h Holter electrocardiogram showed no lethal arrhythmias. Accordingly, neither anticoagulation therapy nor surgical resection of the diverticulum was performed. To the best of our knowledge, ours is the first case of CLVD in a patient with ADPKD. Because gene abnormalities in polycystin coding are mechanistically related to the development of colonic diverticulum and abnormal cyst formation in ADPKD patients, we suspected that CLVD and abnormal cyst formation were related to the same gene abnormality in ADPKD. More case reports, case series studies, and basic research are required to determine whether CLVD in ADPKD is mechanistically associated with abnormal polycystin or just a coincidence.
Collapse
|
24
|
Katsianou MA, Skondra FG, Gargalionis AN, Piperi C, Basdra EK. The role of transient receptor potential polycystin channels in bone diseases. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:246. [PMID: 30069448 DOI: 10.21037/atm.2018.04.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Transient receptor potential (TRP) channels are cation channels which act as molecular sensors that enable cells to detect and respond to a plethora of mechanical and environmental cues. TRPs are involved in various physiological processes, such as mechanosensation, non-inception and thermosensation, while mutations in genes encoding them can lead to pathological conditions, called "channelopathies". The subfamily of transient receptor potential polycystins (TRPPs), Polycystin 1 (PC1, TRPP1) and Polycystin 2 (PC2, TRPP2), act as mechanoreceptors, sensing external mechanical forces, including strain, stretch and fluid shear stress, triggering a cascade of signaling pathways involved in osteoblastogenesis and ultimately bone formation. Both in vitro studies and research on animal models have already identified their implications in bone homeostasis. However, uncertainty veiling the role of polycystins (PCs) in bone disease urges studies to elucidate further their role in this field. Mutations in TRPPs have been related to autosomal polycystic kidney disease (ADKPD) and research groups try to identify their role beyond their well-established contribution in kidney disease. Such an elucidation would be beneficial for identifying signaling pathways where polycystins are involved in bone diseases related to exertion of mechanical forces such as osteoporosis, osteopenia and craniosynostosis. A better understanding of the implications of TRPPs in bone diseases would possibly lay the cornerstone for effective therapeutic schemes.
Collapse
Affiliation(s)
- Maria A Katsianou
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Foteini G Skondra
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios N Gargalionis
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina Piperi
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthimia K Basdra
- Cellular and Molecular Biomechanics Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Ma M, Gallagher AR, Somlo S. Ciliary Mechanisms of Cyst Formation in Polycystic Kidney Disease. Cold Spring Harb Perspect Biol 2017; 9:a028209. [PMID: 28320755 PMCID: PMC5666631 DOI: 10.1101/cshperspect.a028209] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is a disease of defective tissue homeostasis resulting in active remodeling of nephrons and bile ducts to form fluid-filled sacs called cysts. The causal genes PKD1 and PKD2 encode transmembrane proteins polycystin 1 (PC1) and polycystin 2 (PC2), respectively. Together, the polycystins localize to the solitary primary cilium that protrudes from the apical surface of most kidney tubule cells and is thought to function as a privileged compartment that the cell uses for signal integration of sensory inputs. It has been proposed that PC1 and PC2 form a receptor-channel complex that detects external stimuli and transmit a local calcium-mediated signal, which may control a multitude of cellular processes by an as-yet unknown mechanism. Genetic studies using mouse models of cilia and polycystin dysfunction have shown that polycystins regulate an unknown cilia-dependent signal that is normally part of the homeostatic maintenance of nephron structure. ADPKD ensues when this pathway is dysregulated by absence of polycystins from intact cilia, but disruption of cilia also disrupts this signaling mechanism and ameliorates ADPKD even in the absence of polycystins. Understanding the role of cilia and ciliary signaling in ADPKD is challenging, but success will provide saltatory advances in our understanding of how tubule structure is maintained in healthy kidneys and how disruption of polycystin or cilia function leads to the pathological tissue remodeling process underlying ADPKD.
Collapse
Affiliation(s)
- Ming Ma
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029
| | - Anna-Rachel Gallagher
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029
| | - Stefan Somlo
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut 06520-8029
- Department of Genetics, Yale University School of Medicine, New Haven, Connecticut 06520-8029
| |
Collapse
|
26
|
Booij TH, Bange H, Leonhard WN, Yan K, Fokkelman M, Kunnen SJ, Dauwerse JG, Qin Y, van de Water B, van Westen GJP, Peters DJM, Price LS. High-Throughput Phenotypic Screening of Kinase Inhibitors to Identify Drug Targets for Polycystic Kidney Disease. SLAS DISCOVERY 2017. [PMID: 28644734 PMCID: PMC5574491 DOI: 10.1177/2472555217716056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Polycystic kidney disease (PKD) is a prevalent disorder characterized by renal cysts that lead to kidney failure. Various signaling pathways have been targeted to stop disease progression, but most interventions still focus on alleviating PKD-associated symptoms. The mechanistic complexity of the disease, as well as the lack of functional in vitro assays for compound testing, has made drug discovery for PKD challenging. To identify modulators of PKD, Pkd1–/– kidney tubule epithelial cells were applied to a scalable and automated 3D cyst culture model for compound screening, followed by phenotypic profiling to determine compound efficacy. We used this screening platform to screen a library of 273 kinase inhibitors to probe various signaling pathways involved in cyst growth. We show that inhibition of several targets, including aurora kinase, CDK, Chk, IGF-1R, Syk, and mTOR, but, surprisingly, not PI3K, prevented forskolin-induced cyst swelling. Additionally, we show that multiparametric phenotypic classification discriminated potentially undesirable (i.e., cytotoxic) compounds from molecules inducing the desired phenotypic change, greatly facilitating hit selection and validation. Our findings show that a pathophysiologically relevant 3D cyst culture model of PKD coupled to phenotypic profiling can be used to identify potentially therapeutic compounds and predict and validate molecular targets for PKD.
Collapse
Affiliation(s)
- Tijmen H Booij
- 1 Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Hester Bange
- 1 Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Wouter N Leonhard
- 2 Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Kuan Yan
- 3 OcellO B.V., Leiden, Netherlands
| | - Michiel Fokkelman
- 1 Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Steven J Kunnen
- 2 Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | | | - Yu Qin
- 1 Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Bob van de Water
- 1 Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | - Gerard J P van Westen
- 4 Division of Medicinal Chemistry, Leiden Academic Centre for Drug Research, Leiden, Netherlands
| | | | - Leo S Price
- 1 Division of Toxicology, Leiden Academic Centre for Drug Research, Leiden, Netherlands.,3 OcellO B.V., Leiden, Netherlands
| |
Collapse
|
27
|
Lemos FO, Ehrlich BE. Polycystin and calcium signaling in cell death and survival. Cell Calcium 2017; 69:37-45. [PMID: 28601384 DOI: 10.1016/j.ceca.2017.05.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2017] [Revised: 05/18/2017] [Accepted: 05/19/2017] [Indexed: 12/19/2022]
Abstract
Mutations in polycystin-1 (PC1) and polycystin-2 (PC2) result in a commonly occurring genetic disorder, called Autosomal Dominant Polycystic Kidney Disease (ADPKD), that is characterized by the formation and development of kidney cysts. Epithelial cells with loss-of-function of PC1 or PC2 show higher rates of proliferation and apoptosis and reduced autophagy. PC1 is a large multifunctional transmembrane protein that serves as a sensor that is usually found in complex with PC2, a calcium (Ca2+)-permeable cation channel. In addition to decreased Ca2+ signaling, several other cell fate-related pathways are de-regulated in ADPKD, including cAMP, MAPK, Wnt, JAK-STAT, Hippo, Src, and mTOR. In this review we discuss how polycystins regulate cell death and survival, highlighting the complexity of molecular cascades that are involved in ADPKD.
Collapse
Affiliation(s)
- Fernanda O Lemos
- Department of Pharmacology, Yale University, 333 Cedar St, New Haven, CT, 06520, USA
| | - Barbara E Ehrlich
- Department of Pharmacology, Yale University, 333 Cedar St, New Haven, CT, 06520, USA; Department of Cellular and Molecular Physiology, Yale University, 333 Cedar St, New Haven, CT, 06520, USA.
| |
Collapse
|
28
|
Venugopal J, McDermott J, Sanchez G, Sharma M, Barbosa L, Reif GA, Wallace DP, Blanco G. Ouabain promotes partial epithelial to mesenchymal transition (EMT) changes in human autosomal dominant polycystic kidney disease (ADPKD) cells. Exp Cell Res 2017; 355:142-152. [PMID: 28385574 DOI: 10.1016/j.yexcr.2017.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/31/2017] [Accepted: 04/01/2017] [Indexed: 12/13/2022]
Abstract
The hormone ouabain has been shown to enhance the cystic phenotype of autosomal dominant polycystic kidney disease (ADPKD). Among other characteristics, the ADPKD phenotype includes cell de-differentiation and epithelial to mesenchymal transition (EMT). Here, we determined whether physiological concentrations of ouabain induces EMT in human renal epithelial cells from patients with ADPKD. We found that ADPKD cells respond to ouabain with a decrease in expression of the epithelial marker E-cadherin and increase in the expression of the mesenchymal markers N-cadherin, α smooth muscle actin (αSMA) and collagen-I; and the tight junction protein occludin and claudin-1. Other adhesion molecules, such as ZO-1, β-catenin and vinculin were not significantly modified by ouabain. At the cellular level, ouabain stimulated ADPKD cell migration, reduced cell-cell interaction, and the ability of ADPKD cells to form aggregates. Moreover, ouabain increased the transepithelial electrical resistance of ADPKD cell monolayers, suggesting that the paracellular transport pathway was preserved in the cells. These effects of ouabain were not observed in normal human kidney (NHK) cells. Altogether these results show a novel role for ouabain in ADPKD, inducing changes that lead to a partial EMT phenotype in the cells. These effects further support the key role that ouabain has as a factor that promotes the cystic characteristics of ADPKD cells.
Collapse
Affiliation(s)
- Jessica Venugopal
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Jeffrey McDermott
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Gladis Sanchez
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Madhulika Sharma
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Leandro Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Divinopolis, Brazil
| | - Gail A Reif
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Darren P Wallace
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Gustavo Blanco
- Departments of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States; The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, United States.
| |
Collapse
|
29
|
Schenk H, Müller-Deile J, Kinast M, Schiffer M. Disease modeling in genetic kidney diseases: zebrafish. Cell Tissue Res 2017; 369:127-141. [PMID: 28331970 DOI: 10.1007/s00441-017-2593-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/22/2017] [Indexed: 01/07/2023]
Abstract
Growing numbers of translational genomics studies are based on the highly efficient and versatile zebrafish (Danio rerio) vertebrate model. The increasing types of zebrafish models have improved our understanding of inherited kidney diseases, since they not only display pathophysiological changes but also give us the opportunity to develop and test novel treatment options in a high-throughput manner. New paradigms in inherited kidney diseases have been developed on the basis of the distinct genome conservation of approximately 70 % between zebrafish and humans in terms of existing gene orthologs. Several options are available to determine the functional role of a specific gene or gene sets. Permanent genome editing can be induced via complete gene knockout by using the CRISPR/Cas-system, among others, or via transient modification by using various morpholino techniques. Cross-species rescues succeeding knockdown techniques are employed to determine the functional significance of a target gene or a specific mutation. This article summarizes the current techniques and discusses their perspectives.
Collapse
Affiliation(s)
- Heiko Schenk
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Janina Müller-Deile
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Mark Kinast
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA
| | - Mario Schiffer
- Department of Medicine/Nephrology, Hannover Medical School, Hannover, Germany.
- Mount Desert Island Biological Laboratory, Salisbury Cove, Bar Harbor, Me., USA.
| |
Collapse
|
30
|
Abstract
Autosomal dominant polycystic kidney disease is caused by mutation of PKD1 (polycystic kidney disease-1) or PKD2 (polycystic kidney disease-2). PKD1 and PKD2 encode PC1 (polycystin-1) and PC2 (polycystin-2), respectively. In addition, the mutation of cilia-associated proteins is also a recognized major factor of pathogenesis, since PC1 and PC2 are located in primary cilium. Abnormalities of PC1 or PC2 lead to aberrant signaling through downstream pathways, such as the negative growth regulation, G protein activation, and canonical and non-canonical Wnt pathways. According to the "second hit" model, an additional somatic mutation results in the expansion of cyst growth. In this chapter we discuss the genetic mechanisms and signaling pathways involved in ADPKD.
Collapse
|
31
|
Piperi C, Basdra EK. Polycystins and mechanotransduction: From physiology to disease. World J Exp Med 2015; 5:200-205. [PMID: 26618106 PMCID: PMC4655249 DOI: 10.5493/wjem.v5.i4.200] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Revised: 07/21/2015] [Accepted: 09/16/2015] [Indexed: 02/06/2023] Open
Abstract
Polycystins are key mechanosensor proteins able to respond to mechanical forces of external or internal origin. They are widely expressed in primary cilium and plasma membrane of several cell types including kidney, vascular endothelial and smooth muscle cells, osteoblasts and cardiac myocytes modulating their physiology. Interaction of polycystins with diverse ion channels, cell-cell and cell-extracellular matrix junctional proteins implicates them in the regulation of cell structure, mechanical force transmission and mechanotransduction. Their intracellular localization in endoplasmic reticulum further regulates subcellular trafficking and calcium homeostasis, finely-tuning overall cellular mechanosensitivity. Aberrant expression or genetic alterations of polycystins lead to severe structural and mechanosensing abnormalities including cyst formation, deregulated flow sensing, aneurysms, defective bone development and cancer progression, highlighting their vital role in human physiology.
Collapse
|
32
|
Charolidi N, Pirianov G, Torsney E, Pearce S, Laing K, Nohturfft A, Cockerill GW. Pioglitazone Identifies a New Target for Aneurysm Treatment: Role of Egr1 in an Experimental Murine Model of Aortic Aneurysm. J Vasc Res 2015; 52:81-93. [PMID: 26113112 DOI: 10.1159/000430986] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Accepted: 04/26/2015] [Indexed: 11/19/2022] Open
Abstract
Peroxisome proliferator-activated receptor x03B3; agonists have been shown to inhibit angiotensin II (AngII)-induced experimental abdominal aortic aneurysms. Macrophage infiltration to the vascular wall is an early event in this pathology, and therefore we explored the effects of the peroxisome proliferator-activated receptor x03B3; agonist pioglitazone on AngII-treated macrophages. Using microarray-based expression profiling of phorbol ester-stimulated THP-1 cells, we found that a number of aneurysm-related gene changes effected by AngII were modulated following the addition of pioglitazone. Among those genes, polycystic kidney disease 1 (PKD1) was significantly up-regulated (multiple testing corrected p < 0.05). The analysis of the PKD1 proximal promoter revealed a putative early growth response 1 (EGR1) binding site, which was confirmed by chromatin immunoprecipitation (ChIP) and quantitative PCR. Further analysis of publicly available ChIP-sequencing data revealed that this putative binding site overlapped with a conserved EGR1 binding peak present in 5 other cell lines. Quantitative real-time PCR showed that EGR1 suppressed PKD1, while AngII significantly up-regulated PKD1, an effect counteracted by pioglitazone. Conversely, in EGR1 short hairpin RNA lentivirally transduced THP-1 cells, reduced EGR1 led to a significant up-regulation of PKD1, especially after treatment with pioglitazone. In vivo, deficiency of Egr1 in the haematopoietic compartment of mice completely abolished the incidence of CaCl2-induced aneurysm formation.
Collapse
Affiliation(s)
- Nicoletta Charolidi
- Institute of Cardiovascular and Cell Sciences, St. George's, University of London, London, UK
| | | | | | | | | | | | | |
Collapse
|
33
|
Litvinchuk T, Tao Y, Singh R, Vasylyeva TL. A Case of New Familiar Genetic Variant of Autosomal Dominant Polycystic Kidney Disease-2: A Case Study. Front Pediatr 2015; 3:82. [PMID: 26501044 PMCID: PMC4598801 DOI: 10.3389/fped.2015.00082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 09/22/2015] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) is characterized by renal cyst formation due to mutations in genes coding for polycystin-1 [PKD1 (85-90% of cases), on ch 16p13.3] and polycystin-2 [PKD2 (10-15% of cases), on ch 4q13-23] and PKD3 gene (gene unmapped). It is also associated with TSC2/PKD1 contiguous gene syndrome. ADPKD is usually inherited, but new mutations without a family history occur in approximately 10% of the cases. CASE PRESENTATION A 17-year-old boy was followed up for bilateral cystic kidney disease, hypertension, and obesity since he was 13 years old. The diagnosis was an accidental finding during abdominal CT at age 13 to rule out appendicitis. A renal ultrasonogram also demonstrated a multiple bilateral cysts. Because of parental history of bilateral renal cysts, PKD1 and PKD2, genetic testing was ordered. Results showed, PKD2 variant 1:3 bp deletion of TGT; nucleotide position: 1602-1604; codon position: 512-513; mRNA reading frame maintained. The same mutation was later identified in his father. CONCLUSION A smaller number of patients have a defect in the PKD2 locus on chromosome 4 (resulting in PKD2 disease). There are no known published cases on this familiar genetic variant of ADPKD-2 cystic kidney disease. In this case, the disease is present unusually early in life.
Collapse
Affiliation(s)
- Tetiana Litvinchuk
- Department of Pediatrics, Texas Tech Health Sciences Center , Amarillo, TX , USA
| | - Yunxia Tao
- Department of Internal Medicine, Texas Tech Health Sciences Center , Amarillo, TX , USA
| | - Ruchi Singh
- Department of Pediatrics, Texas Tech Health Sciences Center , Amarillo, TX , USA
| | - Tetyana L Vasylyeva
- Department of Pediatrics, Texas Tech Health Sciences Center , Amarillo, TX , USA
| |
Collapse
|
34
|
Cai Y, Fedeles SV, Dong K, Anyatonwu G, Onoe T, Mitobe M, Gao JD, Okuhara D, Tian X, Gallagher AR, Tang Z, Xie X, Lalioti MD, Lee AH, Ehrlich BE, Somlo S. Altered trafficking and stability of polycystins underlie polycystic kidney disease. J Clin Invest 2014; 124:5129-44. [PMID: 25365220 DOI: 10.1172/jci67273] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 09/30/2014] [Indexed: 11/17/2022] Open
Abstract
The most severe form of autosomal dominant polycystic kidney disease occurs in patients with mutations in the gene (PKD1) encoding polycystin-1 (PC1). PC1 is a complex polytopic membrane protein expressed in cilia that undergoes autoproteolytic cleavage at a G protein-coupled receptor proteolytic site (GPS). A quarter of PKD1 mutations are missense variants, though it is not clear how these mutations promote disease. Here, we established a cell-based system to evaluate these mutations and determined that GPS cleavage is required for PC1 trafficking to cilia. A common feature among a subset of pathogenic missense mutations is a resulting failure of PC1 to traffic to cilia regardless of GPS cleavage. The application of our system also identified a missense mutation in the gene encoding polycystin-2 (PC2) that prevented this protein from properly trafficking to cilia. Using a Pkd1-BAC recombineering approach, we developed murine models to study the effects of these mutations and confirmed that only the cleaved form of PC1 exits the ER and can rescue the embryonically lethal Pkd1-null mutation. Additionally, steady-state expression levels of the intramembranous COOH-terminal fragment of cleaved PC1 required an intact interaction with PC2. The results of this study demonstrate that PC1 trafficking and expression require GPS cleavage and PC2 interaction, respectively, and provide a framework for functional assays to categorize the effects of missense mutations in polycystins.
Collapse
|
35
|
Liu D, Xiong S, Zhu Z. Imbalance and dysfunction of transient receptor potential channels contribute to the pathogenesis of hypertension. SCIENCE CHINA-LIFE SCIENCES 2014; 57:818-25. [DOI: 10.1007/s11427-014-4713-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/11/2014] [Indexed: 10/24/2022]
|
36
|
Inoue Y, Sohara E, Kobayashi K, Chiga M, Rai T, Ishibashi K, Horie S, Su X, Zhou J, Sasaki S, Uchida S. Aberrant glycosylation and localization of polycystin-1 cause polycystic kidney in an AQP11 knockout model. J Am Soc Nephrol 2014; 25:2789-99. [PMID: 24854278 DOI: 10.1681/asn.2013060614] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We previously reported that disruption of the aquaporin-11 (AQP11) gene in mice resulted in cystogenesis in the kidney. In this study, we aimed to clarify the mechanism of cystogenesis in AQP11(-/-) mice. To enable the analyses of AQP11 at the protein level in vivo, AQP11 BAC transgenic mice (Tg(AQP11)) that express 3×HA-tagged AQP11 protein were generated. This AQP11 localized to the endoplasmic reticulum (ER) of proximal tubule cells in Tg(AQP11) mice and rescued renal cystogenesis in AQP11(-/-) mice. Therefore, we hypothesized that the absence of AQP11 in the ER could result in impaired quality control and aberrant trafficking of polycystin-1 (PC-1) and polycystin-2 (PC-2). Compared with kidneys of wild-type mice, AQP11(-/-) kidneys exhibited increased protein expression levels of PC-1 and decreased protein expression levels of PC-2. Moreover, PC-1 isolated from AQP11(-/-) mice displayed an altered electrophoretic mobility caused by impaired N-glycosylation processing, and density gradient centrifugation of kidney homogenate and in vivo protein biotinylation revealed impaired membrane trafficking of PC-1 in these mice. Finally, we showed that the Pkd1(+/-) background increased the severity of cystogenesis in AQP11(-/-) mouse kidneys, indicating that PC-1 is involved in the mechanism of cystogenesis in AQP11(-/-) mice. Additionally, the primary cilia of proximal tubules were elongated in AQP11(-/-) mice. Taken together, these data show that impaired glycosylation processing and aberrant membrane trafficking of PC-1 in AQP11(-/-) mice could be a key mechanism of cystogenesis in AQP11(-/-) mice.
Collapse
Affiliation(s)
- Yuichi Inoue
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eisei Sohara
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan;
| | - Katsuki Kobayashi
- Division of Molecular Genetics, Clinical Research Center, Chiba-East National Hospital, Chiba, Japan
| | - Motoko Chiga
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatemitsu Rai
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenichi Ishibashi
- Department of Medical Physiology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Shigeo Horie
- Department of Urology, Juntendo University School of Medicine, Tokyo, Japan; and
| | - Xuefeng Su
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jing Zhou
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Sei Sasaki
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shinichi Uchida
- Department of Nephrology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
37
|
DesRochers TM, Palma E, Kaplan DL. Tissue-engineered kidney disease models. Adv Drug Deliv Rev 2014; 69-70:67-80. [PMID: 24361391 DOI: 10.1016/j.addr.2013.12.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 02/08/2023]
Abstract
Renal disease represents a major health problem that often results in end-stage renal failure necessitating dialysis and eventually transplantation. Historically these diseases have been studied with patient observation and screening, animal models, and two-dimensional cell culture. In this review, we focus on recent advances in tissue engineered kidney disease models that have the capacity to compensate for the limitations of traditional modalities. The cells and materials utilized to develop these models are discussed and tissue engineered models of polycystic kidney disease, drug-induced nephrotoxicity, and the glomerulus are examined in detail. The application of these models has the potential to direct future disease treatments and preclinical drug development.
Collapse
|
38
|
Retailleau K, Duprat F. Polycystins and partners: proposed role in mechanosensitivity. J Physiol 2014; 592:2453-71. [PMID: 24687583 DOI: 10.1113/jphysiol.2014.271346] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Mutations of the two polycystins, PC1 and PC2, lead to polycystic kidney disease. Polycystins are able to form complexes with numerous families of proteins that have been suggested to participate in mechanical sensing. The proposed role of polycystins and their partners in the kidney primary cilium is to sense urine flow. A role for polycystins in mechanosensing has also been shown in other cell types such as vascular smooth muscle cells and cardiac myocytes. At the plasma membrane, polycystins interact with diverse ion channels of the TRP family and with stretch-activated channels (Piezos, TREKs). The actin cytoskeleton and its interacting proteins, such as filamin A, have been shown to be essential for these interactions. Numerous proteins involved in cell-cell and cell-extracellular matrix junctions interact with PC1 and/or PC2. These multimeric protein complexes are important for cell structure integrity, the transmission of force, as well as for mechanosensing and mechanotransduction. A group of polycystin partners are also involved in subcellular trafficking mechanisms. Finally, PC1 and especially PC2 interact with elements of the endoplasmic reticulum and are essential components of calcium homeostasis. In conclusion, we propose that both PC1 and PC2 act as conductors to tune the overall cellular mechanosensitivity.
Collapse
Affiliation(s)
- Kevin Retailleau
- CNRS Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne, France
| | - Fabrice Duprat
- CNRS Institute of Molecular and Cellular Pharmacology (IPMC), Valbonne, France
| |
Collapse
|
39
|
Eneman B, Mekahli D, Audrezet MP, Lerut E, Van Damme-Lombaerts R, Van den Heuvel L, Levtchenko E. An unusual presentation of Denys-Drash syndrome due to bigenic disease. Pediatrics 2014; 133:e252-6. [PMID: 24379226 DOI: 10.1542/peds.2013-1524] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
We report a case of Denys-Drash syndrome (DDS) in a 3-month-old girl presenting with bilateral renal cortical cysts mimicking polycystic kidney disease. Genetic analysis revealed a de novo heterozygous missense mutation c.1186G>A (p.Asp396Asn) in the WT1 gene, confirming the diagnosis of DDS. Because multiple renal cysts have never been reported in DDS, we explored several genes responsible for these renal manifestations, such as HNF-1β, PAX2, PKD1, and PKD2. Remarkably, we identified a heterozygous missense variant c.12439A>G (p.Lys4147Glu) in the PKD1 gene. The same variant was found in the patient's mother, who had no renal cysts, and in the grandfather, who had several renal cysts. Mutation prediction programs classified the c.12439A>G variant as being "likely pathogenic." We hypothesize that the severe cystic phenotype in the index patient could be due to the WT1 mutation, enhancing pathogenicity of the "hypomorph" PKD1 allele. A possible role for Wilms tumor suppressor 1 (WT1) in renal cyst development should be considered. From a conceptual point of view, this case shows that an unusual presentation of a known genetic syndrome might point to bigenic inheritance, with unexpected interference of mutated genes causing an uncommon clinical phenotype.
Collapse
Affiliation(s)
- Benedicte Eneman
- Pediatric Nephrology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium.
| | | | | | | | | | | | | |
Collapse
|
40
|
Fedeles S, Gallagher AR. Cell polarity and cystic kidney disease. Pediatr Nephrol 2013; 28:1161-72. [PMID: 23161205 DOI: 10.1007/s00467-012-2337-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/02/2012] [Accepted: 10/02/2012] [Indexed: 10/27/2022]
Abstract
Epithelial cell polarity is essential for organ development; aberrations in this process have been implicated in various diseases, including polycystic kidney disease. Establishment and maintenance of cell polarity is governed by a number of molecular processes and how these processes operate remains an interesting question. Conserved protein complexes guide both apical-basolateral polarity and planar cell polarity. In this review we discuss the recent findings that provide insights into polarity mechanisms and the intriguing crosstalk between apical-basolateral polarity and planar cell polarity, and their relationship to cystic kidney disease.
Collapse
Affiliation(s)
- Sorin Fedeles
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208029, 333 Cedar Street, New Haven, CT 06520-8029, USA
| | | |
Collapse
|
41
|
Rossetti S, Harris PC. The genetics of vascular complications in autosomal dominant polycystic kidney disease (ADPKD). Curr Hypertens Rev 2013; 9:37-43. [PMID: 23971643 PMCID: PMC4047565 DOI: 10.2174/1573402111309010007] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2012] [Revised: 10/16/2012] [Accepted: 11/26/2012] [Indexed: 11/22/2022]
Abstract
The most important extra-renal manifestation of autosomal dominant polycystic kidney disease (ADPKD) in terms of debilitating injury and premature death is the development of intracranial aneurysms (IAs) and other vascular complications, resulting in subarachnoid hemorrhage (SAH). IAs are found at a rate approximately five times higher in ADPKD patients than in the general population and in patients with a family history of SAH/IAs the frequency is elevated further three to five times, indicating the importance of genetic factors in its etiology. Expression of the ADPKD gene products, polycystin-1 (PKD1) and polycystin-2 (PKD2), in vascular smooth muscle and the endothelium, and evidence that reduced levels of these proteins leads to IA development in mouse models, suggests a direct role of these proteins in the vascular disease. PKD1 and PKD2 patients seem equally likely to develop IAs, while patients with mutations to the 5' half of PKD1 may more likely have vascular complications. Genome wide association and candidate studies of multiplex families with IAs without ADPKD have identified a number of genes/proteins that may be risk factors for the development of IAs. These candidate proteins largely have roles in the maintenance and remodeling of the arterial wall of small brain arteries. The development of the genetic methodologies of massively parallel sequencing mean it is now possible to test these and other candidates in ADPKD families with multiplex and singleton IA cases. Identifying strong modifiers of this phenotype will be important for prioritizing patients for presymptomatic screening and interventions.
Collapse
Affiliation(s)
- Sandro Rossetti
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN 5590, USA
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, 200 First Street SW, Rochester, MN 5590, USA
| |
Collapse
|
42
|
Chiha J, Rangan GK, Chapman JR, Thiagalingam A. Autosomal dominant polycystic kidney disease (ADPKD) is associated with coronary arterial dilatation in end-stage renal failure patients. Clin Kidney J 2012; 5:41-3. [PMID: 26069746 PMCID: PMC4400451 DOI: 10.1093/ndtplus/sfr135] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Accepted: 08/30/2011] [Indexed: 11/14/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) can affect several organs in addition to the kidney. There is paucity in the literature on the cardiac manifestations of this disease. This retrospective study aimed to assess whether ADPKD was associated with a larger coronary artery diameter and to evaluate for the presence of coronary artery aneurysm and ectasia. This study shows that subjects with ADPKD and end-stage renal failure have dilatation of coronary arteries independent of traditional coronary risk factors and medication use.
Collapse
Affiliation(s)
- Joseph Chiha
- Department of Cardiology, Westmead Hospital, Sydney, Australia ; Faculty of Medicine, University of Sydney, Sydney, Australia
| | - Gopala K Rangan
- Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Sydney, Australia
| | - Jeremy R Chapman
- Centre for Transplant and Renal Research, Westmead Millennium Institute, University of Sydney at Westmead Hospital, Sydney, Australia
| | - Aravinda Thiagalingam
- Department of Cardiology, Westmead Hospital, Sydney, Australia ; Faculty of Medicine, University of Sydney, Sydney, Australia
| |
Collapse
|
43
|
Polycystin-1 is required for stereocilia structure but not for mechanotransduction in inner ear hair cells. J Neurosci 2011; 31:12241-50. [PMID: 21865467 DOI: 10.1523/jneurosci.6531-10.2011] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The polycystic kidney disease-1 (Pkd1) gene encodes a large transmembrane protein (polycystin-1, or PC-1) that is reported to function as a fluid flow sensor in the kidney. As a member of the transient receptor potential family, PC-1 has also been hypothesized to play a role in the elusive mechanoelectrical transduction (MET) channel in inner ear hair cells. Here, we analyze two independent mouse models of PC-1, a knock-in (KI) mutant line and a hair cell-specific inducible Cre-mediated knock-out line. Both models exhibit normal MET channel function at neonatal ages despite hearing loss and ultrastructural abnormalities of sterecilia that remain properly polarized at adult ages. These findings demonstrate that PC-1 plays an essential role in stereocilia structure and maintenance but not directly in MET channel function or planar cell polarity. We also demonstrate that PC-1 is colocalized with F-actin in hair cell stereocilia in vivo, using a hemagglutinin-tagged PC-1 KI mouse model, and in renal epithelial cell microvilli in vitro. These results not only demonstrate a novel role for PC-1 in the cochlea, but also suggest insight into the development of polycystic kidney disease.
Collapse
|
44
|
The mechanosensory role of primary cilia in vascular hypertension. Int J Vasc Med 2011; 2011:376281. [PMID: 21748021 PMCID: PMC3124932 DOI: 10.1155/2011/376281] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2011] [Revised: 03/22/2011] [Accepted: 04/09/2011] [Indexed: 12/20/2022] Open
Abstract
Local regulation of vascular tone plays an important role in cardiovascular control of blood pressure. Aside from chemical or hormonal regulations, this local homeostasis is highly regulated by fluid-shear stress. It was previously unclear how vascular endothelial cells were able to sense fluid-shear stress. The cellular functions of mechanosensory cilia within vascular system have emerged recently. In particular, hypertension is insidious and remains a continuous problem that evolves during the course of polycystic kidney disease (PKD). The basic and clinical perspectives on primary cilia are discussed with regard to the pathogenesis of hypertension in PKD.
Collapse
|
45
|
Irazabal MV, Huston J, Kubly V, Rossetti S, Sundsbak JL, Hogan MC, Harris PC, Brown RD, Torres VE. Extended follow-up of unruptured intracranial aneurysms detected by presymptomatic screening in patients with autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol 2011; 6:1274-85. [PMID: 21551026 DOI: 10.2215/cjn.09731110] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND OBJECTIVES Autosomal dominant polycystic kidney disease (ADPKD) patients have an increased risk for intracranial aneurysms (IAs). The importance of screening for unruptured IAs (UIAs) depends on their risks for growth and rupture. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS ADPKD patients with UIAs found by presymptomatic screening with magnetic resonance angiography (MRA) during 1989 to 2009 were followed initially at 6 months and annually, and less frequently after demonstration of stability. RESULTS Forty-five saccular aneurysms were detected in 38 patients from 36 families. Most were small (median diameter 3.5 mm) and in the anterior circulation (84%). Median age at diagnosis was 49 years. During cumulative imaging follow-up of 243 years, one de novo UIA was detected and increased in size from 2 to 4.4 mm over 144 months and two UIAs grew from 4.5 to 5.9 mm and 4.7 to 6.2 mm after 69 and 184 months, respectively. Seven patients did not have imaging follow-up. No change was detected in the remaining 28 patients. During cumulative clinical follow-up of 316 years, no aneurysm ruptured. Five patients died from unrelated causes and two were lost to follow-up after 8 and 120 months. Three patients underwent surgical clipping. CONCLUSIONS Most UIAs detected by presymptomatic screening in ADPKD patients are small and in the anterior circulation. Growth and rupture risks are not higher than those of UIAs in the general population. These data support very selective screening for UIAs in ADPKD patients, and widespread screening is not indicated.
Collapse
Affiliation(s)
- Maria V Irazabal
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN 55901, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Polycystic kidney disease is a common genetic disorder in which fluid-filled cysts displace normal renal tubules. Here we focus on autosomal dominant polycystic kidney disease, which is attributable to mutations in the PKD1 and PKD2 genes and which is characterized by perturbations of renal epithelial cell growth control, fluid transport, and morphogenesis. The mechanisms that connect the underlying genetic defects to disease pathogenesis are poorly understood, but their exploration is shedding new light on interesting cell biological processes and suggesting novel therapeutic targets.
Collapse
Affiliation(s)
- Hannah C Chapin
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | |
Collapse
|
47
|
Yu W, Hill WG, Apodaca G, Zeidel ML. Expression and distribution of transient receptor potential (TRP) channels in bladder epithelium. Am J Physiol Renal Physiol 2010; 300:F49-59. [PMID: 20943764 DOI: 10.1152/ajprenal.00349.2010] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The urothelium is proposed to be a sensory tissue that responds to mechanical stress by undergoing dynamic membrane trafficking and neurotransmitter release; however, the molecular basis of this function is poorly understood. Transient receptor potential (TRP) channels are ideal candidates to fulfill such a role as they can sense changes in temperature, osmolarity, and mechanical stimuli, and several are reported to be expressed in the bladder epithelium. However, their complete expression profile is unknown and their cellular localization is largely undefined. We analyzed expression of all 33 TRP family members in mouse bladder and urothelium by RT-PCR and found 22 specifically expressed in the urothelium. Of the latter, 10 were chosen for closer investigation based on their known mechanosensory or membrane trafficking functions in other cell types. Western blots confirmed urothelial expression of TRPC1, TRPC4, TRPV1, TRPV2, TRPV4, TRPM4, TRPM7, TRPML1, and polycystins 1 and 2 (PKD1 and PKD2) proteins. We further defined the cellular and subcellular localization of all 10 TRP channels. TRPV2 and TRPM4 were prominently localized to the umbrella cell apical membrane, while TRPC4 and TRPV4 were identified on their abluminal surfaces. TRPC1, TRPM7, and TRPML1 were localized to the cytoplasm, while PKD1 and PKD2 were expressed on the apical and basolateral membranes of umbrella cells as well as in the cytoplasm. The cellular location of TRPV1 in the bladder has been debated, but colocalization with neuronal marker calcitonin gene-related peptide indicated clearly that it is present on afferent neurons that extend into the urothelium, but may not be expressed by the urothelium itself. These findings are consistent with the hypothesis that the urothelium acts as a sentinel and by expressing multiple TRP channels it is likely it can detect and presumably respond to a diversity of external stimuli and suggest that it plays an important role in urothelial signal transduction.
Collapse
Affiliation(s)
- Weiqun Yu
- Department of Medicine, Renal-Electrolyte Division, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | | | | | | |
Collapse
|
48
|
Halvorson CR, Bremmer MS, Jacobs SC. Polycystic kidney disease: inheritance, pathophysiology, prognosis, and treatment. Int J Nephrol Renovasc Dis 2010; 3:69-83. [PMID: 21694932 PMCID: PMC3108786 DOI: 10.2147/ijnrd.s6939] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2010] [Indexed: 01/09/2023] Open
Abstract
Both autosomal dominant and recessive polycystic kidney disease are conditions with severe associated morbidity and mortality. Recent advances in the understanding of the genetic and molecular pathogenesis of both ADPKD and ARPKD have resulted in new, targeted therapies designed to disrupt cell signaling pathways responsible for the abnormal cell proliferation, dedifferentiation, apoptosis, and fluid secretion characteristic of the disease. Herein we review the current understanding of the pathophysiology of these conditions, as well as the current treatments derived from our understanding of the mechanisms of these diseases.
Collapse
Affiliation(s)
- Christian R Halvorson
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.
| | | | | |
Collapse
|
49
|
Torres VE, Boletta A, Chapman A, Gattone V, Pei Y, Qian Q, Wallace DP, Weimbs T, Wüthrich RP. Prospects for mTOR inhibitor use in patients with polycystic kidney disease and hamartomatous diseases. Clin J Am Soc Nephrol 2010; 5:1312-29. [PMID: 20498248 DOI: 10.2215/cjn.01360210] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Mammalian target of rapamycin (mTOR) is the core component of two complexes, mTORC1 and mTORC2. mTORC1 is inhibited by rapamycin and analogues. mTORC2 is impeded only in some cell types by prolonged exposure to these compounds. mTOR activation is linked to tubular cell proliferation in animal models and human autosomal dominant polycystic kidney disease (ADPKD). mTOR inhibitors impede cell proliferation and cyst growth in polycystic kidney disease (PKD) models. After renal transplantation, two small retrospective studies suggested that mTOR was more effective than calcineurin inhibitor-based immunosuppression in limiting kidney and/or liver enlargement. By inhibiting vascular remodeling, angiogenesis, and fibrogenesis, mTOR inhibitors may attenuate nephroangiosclerosis, cyst growth, and interstitial fibrosis. Thus, they may benefit ADPKD at multiple levels. However, mTOR inhibition is not without risks and side effects, mostly dose-dependent. Under certain conditions, mTOR inhibition interferes with adaptive increases in renal proliferation necessary for recovery from injury. They restrict Akt activation, nitric oxide synthesis, and endothelial cell survival (downstream from mTORC2) and potentially increase the risk for glomerular and peritubular capillary loss, vasospasm, and hypertension. They impair podocyte integrity pathways and may predispose to glomerular injury. Administration of mTOR inhibitors is discontinued because of side effects in up to 40% of transplant recipients. Currently, treatment with mTOR inhibitors should not be recommended to treat ADPKD. Results of ongoing studies must be awaited and patients informed accordingly. If effective, lower dosages than those used to prevent rejection would minimize side effects. Combination therapy with other effective drugs could improve tolerability and results.
Collapse
Affiliation(s)
- Vicente E Torres
- Division of Nephrology, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Jain R, Pan J, Driscoll JA, Wisner JW, Huang T, Gunsten SP, You Y, Brody SL. Temporal relationship between primary and motile ciliogenesis in airway epithelial cells. Am J Respir Cell Mol Biol 2010; 43:731-9. [PMID: 20118219 DOI: 10.1165/rcmb.2009-0328oc] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Cilia are traditionally classified as motile or primary. Motile cilia are restricted to specific populations of well-differentiated epithelial cells, including those in the airway, brain ventricles, and oviducts. Primary cilia are nonmotile, solitary structures that are present in many cell types, and often have sensory functions such as in the retina and renal tubules. Primary cilia were also implicated in the regulation of fundamental processes in development. Rare depictions of primary cilia in embryonic airways led us to hypothesize that primary cilia in airway cells are temporally related to motile ciliogenesis. We identified primary cilia in undifferentiated, cultured airway epithelial cells from mice and humans and in developing lungs. The solitary cilia in the airways express proteins considered unique to primary cilia, including polycystin-1 and polycystin-2. A temporal analysis of airway epithelial cell differentiation showed that cells with primary cilia acquire markers of motile ciliogenesis, suggesting that motile ciliated cells originate from primary ciliated cells. Whereas motile ciliogenesis requires Foxj1, primary ciliogenesis does not, and the expression of Foxj1 was associated with a loss of primary cilia, just before the appearance of motile cilia. Primary cilia were not found in well-differentiated airway epithelial cells. However, after injury, they appear in the luminal layer of epithelium and in basal cells. The transient nature of primary cilia, together with the temporal and spatial patterns of expression in the development and repair of airway epithelium, suggests a critical role of primary cilia in determining outcomes during airway epithelial cell differentiation.
Collapse
Affiliation(s)
- Raksha Jain
- Division of Pulmonary and Critical Care, Department of Internal Medicine, Washington University School of Medicine, 660 South Euclid Ave., St. Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|