101
|
Sicking M, Lang S, Bochen F, Roos A, Drenth JPH, Zakaria M, Zimmermann R, Linxweiler M. Complexity and Specificity of Sec61-Channelopathies: Human Diseases Affecting Gating of the Sec61 Complex. Cells 2021; 10:1036. [PMID: 33925740 PMCID: PMC8147068 DOI: 10.3390/cells10051036] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 12/14/2022] Open
Abstract
The rough endoplasmic reticulum (ER) of nucleated human cells has crucial functions in protein biogenesis, calcium (Ca2+) homeostasis, and signal transduction. Among the roughly one hundred components, which are involved in protein import and protein folding or assembly, two components stand out: The Sec61 complex and BiP. The Sec61 complex in the ER membrane represents the major entry point for precursor polypeptides into the membrane or lumen of the ER and provides a conduit for Ca2+ ions from the ER lumen to the cytosol. The second component, the Hsp70-type molecular chaperone immunoglobulin heavy chain binding protein, short BiP, plays central roles in protein folding and assembly (hence its name), protein import, cellular Ca2+ homeostasis, and various intracellular signal transduction pathways. For the purpose of this review, we focus on these two components, their relevant allosteric effectors and on the question of how their respective functional cycles are linked in order to reconcile the apparently contradictory features of the ER membrane, selective permeability for precursor polypeptides, and impermeability for Ca2+. The key issues are that the Sec61 complex exists in two conformations: An open and a closed state that are in a dynamic equilibrium with each other, and that BiP contributes to its gating in both directions in cooperation with different co-chaperones. While the open Sec61 complex forms an aqueous polypeptide-conducting- and transiently Ca2+-permeable channel, the closed complex is impermeable even to Ca2+. Therefore, we discuss the human hereditary and tumor diseases that are linked to Sec61 channel gating, termed Sec61-channelopathies, as disturbances of selective polypeptide-impermeability and/or aberrant Ca2+-permeability.
Collapse
Affiliation(s)
- Mark Sicking
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Sven Lang
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Florian Bochen
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, D-66421 Homburg, Germany; (F.B.); (M.L.)
| | - Andreas Roos
- Department of Neuropediatrics, Essen University Hospital, D-45147 Essen, Germany;
| | - Joost P. H. Drenth
- Department of Molecular Gastroenterology and Hepatology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Muhammad Zakaria
- Department of Genetics, Hazara University, Mansehra 21300, Pakistan;
| | - Richard Zimmermann
- Department of Medical Biochemistry & Molecular Biology, Saarland University, D-66421 Homburg, Germany;
| | - Maximilian Linxweiler
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, D-66421 Homburg, Germany; (F.B.); (M.L.)
| |
Collapse
|
102
|
Cabrita I, Talbi K, Kunzelmann K, Schreiber R. Loss of PKD1 and PKD2 share common effects on intracellular Ca 2+ signaling. Cell Calcium 2021; 97:102413. [PMID: 33915319 DOI: 10.1016/j.ceca.2021.102413] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 04/17/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022]
Abstract
In polycystic kidney disease (PKD) multiple bilateral renal cysts gradually enlarge causing a decline in renal function. Transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and TMEM16A (anoctamin 1) drive cyst enlargement. We demonstrated recently that a loss of PKD1 increases expression and function of TMEM16A in murine kidneys and in mouse M1 collecting duct cells. The data demonstrated that TMEM16A contributes essentially to cyst growth by upregulating intracellular Ca2+ signaling. Enhanced expression of TMEM16A and Ca2+ signaling increased both cell proliferation and fluid secretion, which suggested inhibition of TMEM16A as a novel therapy in ADPKD. About 15 % of all ADPKD cases are caused by mutations in PKD2. To analyze the effects of loss of function of PKD2 on Ca2+ signaling, we knocked-down Pkd2 in mouse primary renal epithelial cells in the present study, using viral transfection of shRNA. Unlike in Pkd1-/- cells, knockdown of PKD2 lowered basal Ca2+ and augmented store-operated Ca2+ entry, which was both independent of TMEM16A. However, disease causing purinergic Ca2+ store release was enhanced, similar to that observed in Pkd1-/- renal epithelial cells. The present data suggest pharmacological inhibition of TMEM16A as a treatment in ADPKD caused by mutations in both PKD1 and PKD2.
Collapse
Affiliation(s)
- Ines Cabrita
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Khaoula Talbi
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| | - Karl Kunzelmann
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany.
| | - Rainer Schreiber
- Institut für Physiologie, Universität Regensburg, Universitätsstraße 31, D-93053 Regensburg, Germany
| |
Collapse
|
103
|
Millet-Boureima C, He S, Le TBU, Gamberi C. Modeling Neoplastic Growth in Renal Cell Carcinoma and Polycystic Kidney Disease. Int J Mol Sci 2021; 22:3918. [PMID: 33920158 PMCID: PMC8070407 DOI: 10.3390/ijms22083918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) and autosomal dominant polycystic kidney disease (ADPKD) share several characteristics, including neoplastic cell growth, kidney cysts, and limited therapeutics. As well, both exhibit impaired vasculature and compensatory VEGF activation of angiogenesis. The PI3K/AKT/mTOR and Ras/Raf/ERK pathways play important roles in regulating cystic and tumor cell proliferation and growth. Both RCC and ADPKD result in hypoxia, where HIF-α signaling is activated in response to oxygen deprivation. Primary cilia and altered cell metabolism may play a role in disease progression. Non-coding RNAs may regulate RCC carcinogenesis and ADPKD through their varied effects. Drosophila exhibits remarkable conservation of the pathways involved in RCC and ADPKD. Here, we review the progress towards understanding disease mechanisms, partially overlapping cellular and molecular dysfunctions in RCC and ADPKD and reflect on the potential for the agile Drosophila genetic model to accelerate discovery science, address unresolved mechanistic aspects of these diseases, and perform rapid pharmacological screens.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Thi Bich Uyen Le
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore 119228, Singapore
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29528-6054, USA
| |
Collapse
|
104
|
Lemos FO, Bultynck G, Parys JB. A comprehensive overview of the complex world of the endo- and sarcoplasmic reticulum Ca 2+-leak channels. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119020. [PMID: 33798602 DOI: 10.1016/j.bbamcr.2021.119020] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 12/11/2022]
Abstract
Inside cells, the endoplasmic reticulum (ER) forms the largest Ca2+ store. Ca2+ is actively pumped by the SERCA pumps in the ER, where intraluminal Ca2+-binding proteins enable the accumulation of large amount of Ca2+. IP3 receptors and the ryanodine receptors mediate the release of Ca2+ in a controlled way, thereby evoking complex spatio-temporal signals in the cell. The steady state Ca2+ concentration in the ER of about 500 μM results from the balance between SERCA-mediated Ca2+ uptake and the passive leakage of Ca2+. The passive Ca2+ leak from the ER is often ignored, but can play an important physiological role, depending on the cellular context. Moreover, excessive Ca2+ leakage significantly lowers the amount of Ca2+ stored in the ER compared to normal conditions, thereby limiting the possibility to evoke Ca2+ signals and/or causing ER stress, leading to pathological consequences. The so-called Ca2+-leak channels responsible for Ca2+ leakage from the ER are however still not well understood, despite over 20 different proteins have been proposed to contribute to it. This review has the aim to critically evaluate the available evidence about the various channels potentially involved and to draw conclusions about their relative importance.
Collapse
Affiliation(s)
- Fernanda O Lemos
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Geert Bultynck
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium
| | - Jan B Parys
- KU Leuven, Laboratory of Molecular and Cellular Signaling, Department of Cellular and Molecular Medicine and Leuven Kanker Instituut, Campus Gasthuisberg O/N-1 B-802, Herestraat 49, B-3000 Leuven, Belgium.
| |
Collapse
|
105
|
Balancing ER-Mitochondrial Ca 2+ Fluxes in Health and Disease. Trends Cell Biol 2021; 31:598-612. [PMID: 33678551 DOI: 10.1016/j.tcb.2021.02.003] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023]
Abstract
Organelles cooperate with each other to control cellular homeostasis and cell functions by forming close connections through membrane contact sites. Important contacts are present between the endoplasmic reticulum (ER), the main intracellular Ca2+-storage organelle, and the mitochondria, the organelle responsible not only for the majority of cellular ATP production but also for switching on cell death processes. Several Ca2+-transport systems focalize at these contact sites, thereby enabling the efficient transmission of Ca2+ signals from the ER toward mitochondria. This provides tight control of mitochondrial functions at the microdomain level. Here, we discuss how ER-mitochondrial Ca2+ transfers support cell function and how their dysregulation underlies, drives, or contributes to pathogenesis and pathophysiology, with a major focus on cancer and neurodegeneration but also with attention to other diseases such as diabetes and rare genetic diseases.
Collapse
|
106
|
Automatic semantic segmentation of kidney cysts in MR images of patients affected by autosomal-dominant polycystic kidney disease. Abdom Radiol (NY) 2021; 46:1053-1061. [PMID: 32940759 PMCID: PMC7940295 DOI: 10.1007/s00261-020-02748-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/26/2020] [Accepted: 09/03/2020] [Indexed: 12/22/2022]
Abstract
PURPOSE For patients affected by autosomal-dominant polycystic kidney disease (ADPKD), successful differentiation of cysts is useful for automatic classification of patient phenotypes, clinical decision-making, and disease progression. The objective was to develop and evaluate a fully automated semantic segmentation method to differentiate and analyze renal cysts in patients with ADPKD. METHODS An automated deep learning approach using a convolutional neural network was trained, validated, and tested on a set of 60 MR T2-weighted images. A three-fold cross-validation approach was used to train three models on distinct training and validation sets (n = 40). An ensemble model was then built and tested on the hold out cases (n = 20), with each of the cases compared to manual segmentations performed by two readers. Segmentation agreement between readers and the automated method was assessed. RESULTS The automated approach was found to perform at the level of interobserver variability. The automated approach had a Dice coefficient (mean ± standard deviation) of 0.86 ± 0.10 vs Reader-1 and 0.84 ± 0.11 vs. Reader-2. Interobserver Dice was 0.86 ± 0.08. In terms of total cyst volume (TCV), the automated approach had a percent difference of 3.9 ± 19.1% vs Reader-1 and 8.0 ± 24.1% vs Reader-2, whereas interobserver variability was - 2.0 ± 16.4%. CONCLUSION This study developed and validated a fully automated approach for performing semantic segmentation of kidney cysts in MR images of patients affected by ADPKD. This approach will be useful for exploring additional imaging biomarkers of ADPKD and automatically classifying phenotypes.
Collapse
|
107
|
Linn E, Ghanem L, Bhakta H, Greer C, Avella M. Genes Regulating Spermatogenesis and Sperm Function Associated With Rare Disorders. Front Cell Dev Biol 2021; 9:634536. [PMID: 33665191 PMCID: PMC7921155 DOI: 10.3389/fcell.2021.634536] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 01/20/2021] [Indexed: 12/26/2022] Open
Abstract
Spermatogenesis is a cell differentiation process that ensures the production of fertilizing sperm, which ultimately fuse with an egg to form a zygote. Normal spermatogenesis relies on Sertoli cells, which preserve cell junctions while providing nutrients for mitosis and meiosis of male germ cells. Several genes regulate normal spermatogenesis, some of which are not exclusively expressed in the testis and control multiple physiological processes in an organism. Loss-of-function mutations in some of these genes result in spermatogenesis and sperm functionality defects, potentially leading to the insurgence of rare genetic disorders. To identify genetic intersections between spermatogenesis and rare diseases, we screened public archives of human genetic conditions available on the Genetic and Rare Diseases Information Center (GARD), the Online Mendelian Inheritance in Man (OMIM), and the Clinical Variant (ClinVar), and after an extensive literature search, we identified 22 distinct genes associated with 21 rare genetic conditions and defective spermatogenesis or sperm function. These protein-coding genes regulate Sertoli cell development and function during spermatogenesis, checkpoint signaling pathways at meiosis, cellular organization and shape definition during spermiogenesis, sperm motility, and capacitation at fertilization. A number of these genes regulate folliculogenesis and oogenesis as well. For each gene, we review the genotype–phenotype association together with associative or causative polymorphisms in humans, and provide a description of the shared molecular mechanisms that regulate gametogenesis and fertilization obtained in transgenic animal models.
Collapse
Affiliation(s)
- Emma Linn
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Lillian Ghanem
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Hanisha Bhakta
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Cory Greer
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| | - Matteo Avella
- Department of Biological Science, College of Engineering and Natural Sciences, University of Tulsa, Tulsa, OK, United States
| |
Collapse
|
108
|
Ala M, Mohammad Jafari R, Hajiabbasi A, Dehpour AR. Aquaporins and diseases pathogenesis: From trivial to undeniable involvements, a disease-based point of view. J Cell Physiol 2021; 236:6115-6135. [PMID: 33559160 DOI: 10.1002/jcp.30318] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 01/01/2023]
Abstract
Aquaporins (AQPs), as transmembrane proteins, were primarily identified as water channels with the ability of regulating the transmission of water, glycerol, urea, and other small-sized molecules. The classic view of AQPs involvement in therapeutic plan restricted them and their regulators into managing only a narrow spectrum of the diseases such as diabetes insipidus and the syndrome of inappropriate ADH secretion. However, further investigations performed, especially in the third millennium, has found that their cooperation in water transmission control can be manipulated to handle other burden-imposing diseases such as cirrhosis, heart failure, Meniere's disease, cancer, bullous pemphigoid, eczema, and Sjögren's syndrome.
Collapse
Affiliation(s)
- Moein Ala
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Razieh Mohammad Jafari
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Asghar Hajiabbasi
- Guilan Rheumatology Research Center, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Ahmad Reza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
109
|
Little RB, Norris DP. Right, left and cilia: How asymmetry is established. Semin Cell Dev Biol 2021; 110:11-18. [PMID: 32571625 DOI: 10.1016/j.semcdb.2020.06.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/03/2020] [Accepted: 06/04/2020] [Indexed: 12/13/2022]
Abstract
The initial breaking of left-right (L-R) symmetry in the embryo is controlled by a motile-cilia-driven leftward fluid flow in the left-right organiser (LRO), resulting in L-R asymmetric gene expression flanking the LRO. Ultimately this results in left- but not right-sided activation of the Nodal-Pitx2 pathway in more lateral tissues. While aspects of the initial breaking event clearly vary between vertebrates, events in the Lateral Plate Mesoderm (LPM) are conserved through the vertebrate lineage. Evidence from model systems and humans highlights the role of cilia both in the initial symmetry breaking and in the ability of more lateral tissues to exhibit asymmetric gene expression. In this review we concentrate on the process of L-R determination in mouse and humans.
Collapse
Affiliation(s)
- Rosie B Little
- MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK
| | - Dominic P Norris
- MRC Harwell Institute, Harwell Campus, Oxfordshire, OX11 0RD, UK.
| |
Collapse
|
110
|
Corkins ME, Krneta-Stankic V, Kloc M, Miller RK. Aquatic models of human ciliary diseases. Genesis 2021; 59:e23410. [PMID: 33496382 PMCID: PMC8593908 DOI: 10.1002/dvg.23410] [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: 11/09/2020] [Revised: 01/08/2021] [Accepted: 01/09/2021] [Indexed: 11/06/2022]
Abstract
Cilia are microtubule-based structures that either transmit information into the cell or move fluid outside of the cell. There are many human diseases that arise from malfunctioning cilia. Although mammalian models provide vital insights into the underlying pathology of these diseases, aquatic organisms such as Xenopus and zebrafish provide valuable tools to help screen and dissect out the underlying causes of these diseases. In this review we focus on recent studies that identify or describe different types of human ciliopathies and outline how aquatic organisms have aided our understanding of these diseases.
Collapse
Affiliation(s)
- Mark E. Corkins
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston Texas 77030
| | - Vanja Krneta-Stankic
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genes & Development, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics & Epigenetics, Houston, Texas 77030
| | - Malgorzata Kloc
- Houston Methodist, Research Institute, Houston Texas 77030
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston Texas 77030
| | - Rachel K. Miller
- Department of Pediatrics, Pediatric Research Center, UTHealth McGovern Medical School, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics & Epigenetics, Houston, Texas 77030
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston Texas 77030
- MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Biochemistry & Cell Biology, Houston Texas 77030
| |
Collapse
|
111
|
Sparapani S, Millet-Boureima C, Oliver J, Mu K, Hadavi P, Kalostian T, Ali N, Avelar CM, Bardies M, Barrow B, Benedikt M, Biancardi G, Bindra R, Bui L, Chihab Z, Cossitt A, Costa J, Daigneault T, Dault J, Davidson I, Dias J, Dufour E, El-Khoury S, Farhangdoost N, Forget A, Fox A, Gebrael M, Gentile MC, Geraci O, Gnanapragasam A, Gomah E, Haber E, Hamel C, Iyanker T, Kalantzis C, Kamali S, Kassardjian E, Kontos HK, Le TBU, LoScerbo D, Low YF, Mac Rae D, Maurer F, Mazhar S, Nguyen A, Nguyen-Duong K, Osborne-Laroche C, Park HW, Parolin E, Paul-Cole K, Peer LS, Philippon M, Plaisir CA, Porras Marroquin J, Prasad S, Ramsarun R, Razzaq S, Rhainds S, Robin D, Scartozzi R, Singh D, Fard SS, Soroko M, Soroori Motlagh N, Stern K, Toro L, Toure MW, Tran-Huynh S, Trépanier-Chicoine S, Waddingham C, Weekes AJ, Wisniewski A, Gamberi C. The Biology of Vasopressin. Biomedicines 2021; 9:89. [PMID: 33477721 PMCID: PMC7832310 DOI: 10.3390/biomedicines9010089] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/29/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Vasopressins are evolutionarily conserved peptide hormones. Mammalian vasopressin functions systemically as an antidiuretic and regulator of blood and cardiac flow essential for adapting to terrestrial environments. Moreover, vasopressin acts centrally as a neurohormone involved in social and parental behavior and stress response. Vasopressin synthesis in several cell types, storage in intracellular vesicles, and release in response to physiological stimuli are highly regulated and mediated by three distinct G protein coupled receptors. Other receptors may bind or cross-bind vasopressin. Vasopressin is regulated spatially and temporally through transcriptional and post-transcriptional mechanisms, sex, tissue, and cell-specific receptor expression. Anomalies of vasopressin signaling have been observed in polycystic kidney disease, chronic heart failure, and neuropsychiatric conditions. Growing knowledge of the central biological roles of vasopressin has enabled pharmacological advances to treat these conditions by targeting defective systemic or central pathways utilizing specific agonists and antagonists.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada; (S.S.); (C.M.-B.); (J.O.); (K.M.); (P.H.); (T.K.); (N.A.); (C.M.A.); (M.B.); (B.B.); (M.B.); (G.B.); (R.B.); (L.B.); (Z.C.); (A.C.); (J.C.); (T.D.); (J.D.); (I.D.); (J.D.); (E.D.); (S.E.-K.); (N.F.); (A.F.); (A.F.); (M.G.); (M.C.G.); (O.G.); (A.G.); (E.G.); (E.H.); (C.H.); (T.I.); (C.K.); (S.K.); (E.K.); (H.K.K.); (T.B.U.L.); (D.L.); (Y.F.L.); (D.M.R.); (F.M.); (S.M.); (A.N.); (K.N.-D.); (C.O.-L.); (H.W.P.); (E.P.); (K.P.-C.); (L.S.P.); (M.P.); (C.-A.P.); (J.P.M.); (S.P.); (R.R.); (S.R.); (S.R.); (D.R.); (R.S.); (D.S.); (S.S.F.); (M.S.); (N.S.M.); (K.S.); (L.T.); (M.W.T.); (S.T.-H.); (S.T.-C.); (C.W.); (A.J.W.); (A.W.)
| |
Collapse
|
112
|
Cansever HN, Sari F, Cevikol C, Cetinkaya R, Süleymanlar G, Ersoy F. Serum uromodulin levels, MR imaging findings, and their relationship with eGFR-based CKD staging in ADPKD patients. Int Urol Nephrol 2021; 53:1383-1389. [PMID: 33389516 DOI: 10.1007/s11255-020-02730-5] [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: 04/06/2020] [Accepted: 12/02/2020] [Indexed: 10/22/2022]
Abstract
PURPOSE Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease that may progress to end-stage renal disease, characterized by increased kidney volume due to cystic formations. In this study, we aimed to investigate the relationship between serum uromodulin levels, total kidney volume and estimated glomerular filtration rate (eGFR) in patients with ADPKD. METHODS This study included a total of 54 ADPKD patients and 18 healthy volunteers (control group). Total kidney volumes were calculated through magnetic resonance images using ellipsoid method. Serum uromodulin measurements were measured using an ELISA method. RESULTS Serum uromodulin levels were lower in patients compared with the control group (2.47 ± 0.16 vs 2.6 ± 0.28, p = 0.021). There was no significant difference in uromodulin values among the patients in chronic kidney disease (CKD) stages 1-2, 3 and 4-5. TKV measurements of CKD stage 4-5 patients were significantly higher than the stage 1-2 patients (p = 0.015). A negative correlation was observed between TKV and eGFR (r = - 0.433, p = 0.001). A positive correlation was observed between uromodulin and eGFR (r = 0.274, p = 0.02). When the serum levels of uromodulin and the level of eGFR were evaluated using simple linear regression analysis, R2 value was found to be 0.075, suggesting that 7.5% change in serum uromodulin values corresponds with the change in eGFR value. CONCLUSION These findings are consistent with previous studies that reported that serum uromodulin may be a good biomarker for demonstrating renal function in the early stages of CKD, before eGFR levels deteriorate. Serum uromodulin level may be useful in demonstrating renal functions in the follow-up of individuals with ADPKD.
Collapse
Affiliation(s)
- Hale Nur Cansever
- Division of Nephrology, Department of Medicine, Akdeniz University Hospital, Dumlupinar Bulvari, Konyaaltı, 07070, Antalya, Turkey
| | - Funda Sari
- Division of Nephrology, Department of Medicine, Akdeniz University Hospital, Dumlupinar Bulvari, Konyaaltı, 07070, Antalya, Turkey.
| | - Can Cevikol
- Division of Nephrology, Department of Medicine, Akdeniz University Hospital, Dumlupinar Bulvari, Konyaaltı, 07070, Antalya, Turkey
| | - Ramazan Cetinkaya
- Division of Nephrology, Department of Medicine, Akdeniz University Hospital, Dumlupinar Bulvari, Konyaaltı, 07070, Antalya, Turkey
| | - Gultekin Süleymanlar
- Division of Nephrology, Department of Medicine, Akdeniz University Hospital, Dumlupinar Bulvari, Konyaaltı, 07070, Antalya, Turkey
| | - Fevzi Ersoy
- Division of Nephrology, Department of Medicine, Akdeniz University Hospital, Dumlupinar Bulvari, Konyaaltı, 07070, Antalya, Turkey
| |
Collapse
|
113
|
Seliger SL, Watnick T, Althouse AD, Perrone RD, Abebe KZ, Hallows KR, Miskulin DC, Bae KT. Baseline Characteristics and Patient-Reported Outcomes of ADPKD Patients in the Multicenter TAME-PKD Clinical Trial. KIDNEY360 2020; 1:1363-1372. [PMID: 33768205 PMCID: PMC7990324 DOI: 10.34067/kid.0004002020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/22/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) has been associated with metabolic disturbances characterized by downregulation of AMP-activated protein kinase (AMPK), a critical sensor of the cellular energy status. Therapeutic activation of AMPK by metformin could inhibit cyst enlargement by inhibition of both the mammalian target of rapamycin pathway and fluid secretion via the CFTR chloride channel. METHODS We designed a phase-2, randomized, placebo-controlled, clinical trial to assess the safety, tolerability, and efficacy of metformin on total kidney volume in adults without diabetes (age 18-60 years) with ADPKD and eGFR of ≥50 ml/min per 1.73 m2. There were no eligibility criteria relating to kidney volume. In addition to demographics and clinical/family history, baseline parameters included eGFR, total kidney and liver volumes measured by MRI, and patient-reported outcomes were ascertained by the Medical Outcomes Study Short Form-36, the Gastrointestinal Safety Rating Scale, and the HALT-PKD pain questionnaire. RESULTS We successfully randomized 97 participants recruited from two university-based clinical sites in Baltimore and Boston. The mean age of participants was 41.9 years, 72% were female, and 94% of participants were White. The majority of study participants had early stage disease, with a mean eGFR of 86.8±19.0 ml/min per 1.73 m2. Approximately half of the study participants (48%) were classified as high risk for progression (Mayo imaging classes 1C, 1D, or 1E). There was no correlation between kidney and/or liver size and health-related quality of life (HRQoL) or gastrointestinal symptom severity. CONCLUSIONS We report successful recruitment in this ongoing, novel, clinical trial of metformin in ADPKD, with a study sample comprising patients with early stage disease and nearly a half of participants considered at high estimated risk for progression. Participants reported a low gastrointestinal symptom burden at baseline, and HRQoL similar to that of the general population, with no differences in symptoms or HRQoL related to organomegaly. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Metformin as a Novel Therapy for Autosomal Dominant Polycystic Kidney Disease (TAME), NCT02656017.
Collapse
Affiliation(s)
- Stephen L. Seliger
- Department of Medicine, Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Terry Watnick
- Department of Medicine, Division of Nephrology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Andrew D. Althouse
- Department of Medicine, Division of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ronald D. Perrone
- Department of Medicine, Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
| | - Kaleab Z. Abebe
- Department of Medicine, Division of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kenneth R. Hallows
- Department of Medicine, Division of Nephrology and Hypertension, University of Southern California Keck School of Medicine, Los Angeles, California
| | - Dana C. Miskulin
- Department of Medicine, Division of Internal Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kyongtae T. Bae
- Department of Medicine, Division of Nephrology, Tufts Medical Center, Boston, Massachusetts
- Department of RadiologyDepartment of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
114
|
Srivastava SP, Kanasaki K, Goodwin JE. Loss of Mitochondrial Control Impacts Renal Health. Front Pharmacol 2020; 11:543973. [PMID: 33362536 PMCID: PMC7756079 DOI: 10.3389/fphar.2020.543973] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022] Open
Abstract
Disruption of mitochondrial biosynthesis or dynamics, or loss of control over mitochondrial regulation leads to a significant alteration in fuel preference and metabolic shifts that potentially affect the health of kidney cells. Mitochondria regulate metabolic networks which affect multiple cellular processes. Indeed, mitochondria have established themselves as therapeutic targets in several diseases. The importance of mitochondria in regulating the pathogenesis of several diseases has been recognized, however, there is limited understanding of mitochondrial biology in the kidney. This review provides an overview of mitochondrial dysfunction in kidney diseases. We describe the importance of mitochondria and mitochondrial sirtuins in the regulation of renal metabolic shifts in diverse cells types, and review this loss of control leads to increased cell-to-cell transdifferentiation processes and myofibroblast-metabolic shifts, which affect the pathophysiology of several kidney diseases. In addition, we examine mitochondrial-targeted therapeutic agents that offer potential leads in combating kidney diseases.
Collapse
Affiliation(s)
- Swayam Prakash Srivastava
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Keizo Kanasaki
- Internal Medicine 1, Shimane University Faculty of Medicine, Izumo, Japan
| | - Julie E. Goodwin
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
- Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| |
Collapse
|
115
|
Ordikhani F, Kasinath V, Uehara M, Akbarzadeh A, Yilmam OA, Dai L, Aksu H, Jung S, Jiang L, Li X, Zhao J, Bahmani B, Ichimura T, Fiorina P, Annabi N, Abdi R. Selective Trafficking of Light Chain-Conjugated Nanoparticles to the Kidney and Renal Cell Carcinoma. NANO TODAY 2020; 35:100990. [PMID: 33244320 PMCID: PMC7685247 DOI: 10.1016/j.nantod.2020.100990] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Specific delivery platforms for drugs to the kidney and diagnostic agents to renal cell carcinoma (RCC) constitute urgent but unfulfilled clinical needs. To address these challenges, we engineered nanocarriers that interact selectively for the first time with proximal tubule epithelial cells (PTECs) in the kidney and with RCC through the interplay between lambda light chains (LCs) attached to PEGylated polylactic-co-glycolic acid (PLGA) nanoparticles and the membrane protein megalin. Systemic administration of these light chain-conjugated nanoparticles (LC-NPs) to mice resulted in their specific retention by megalin-expressing PTECs for seven days. Repetitive dosing of LC-NPs demonstrated no renal toxicity. LC-NPs also localized selectively to megalin-expressing RCC tumors in mice. Moreover, we confirmed that both the primary tumor and lymph node metastases of human RCC express megalin, reinforcing the potential of LC-NPs for clinical use. Thus, LC-NPs can contribute potentially to improving the management of both non-oncologic and oncologic renal disorders.
Collapse
Affiliation(s)
- Farideh Ordikhani
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Vivek Kasinath
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Mayuko Uehara
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Aram Akbarzadeh
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Osman A Yilmam
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Li Dai
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Hamza Aksu
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Sungwook Jung
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Liwei Jiang
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Xiaofei Li
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Jing Zhao
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Baharak Bahmani
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Takaharu Ichimura
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Paolo Fiorina
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Nephrology, Boston Children’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Nasim Annabi
- Chemical and Biomolecular Engineering Department and Center for Minimally Invasive Therapeutics (C-MIT), California NanoSystems Institute (CNSI), University of California - Los Angeles, Los Angeles, CA, USA
| | - Reza Abdi
- Transplantation Research Center, Division of Renal Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
116
|
Kalatharan V, Welk B, Nash DM, McArthur E, Slater J, Sarma S, Pei Y, Garg AX. Complications in Patients With Autosomal Dominant Polycystic Kidney Disease Undergoing Ureteroscopy: A Cohort Study. Can J Kidney Health Dis 2020; 7:2054358120972830. [PMID: 33282326 PMCID: PMC7691941 DOI: 10.1177/2054358120972830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 09/30/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Ureteroscopy is a minimally invasive treatment option for upper tract stones. The distorted kidney anatomy in patients with autosomal dominant polycystic kidney disease (ADPKD) may place them at higher risk for ureteroscopic complications. OBJECTIVE To compare the 30-day risk of ureteroscopic complications between patients with and without ADPKD. DESIGN Retrospective cohort study. SETTING Ontario, Canada. PATIENTS Seventy three patients with ADPKD and 81 445 patients without ADPKD who underwent ureteroscopy for upper urinary tract stones between April 1, 2002, and March 1, 2018. MEASUREMENTS A 30-day risk of (1) hospital presentation with ureteroscopic complications (which was a composite outcome of either emergency department visit or hospital admission with acute kidney injury, urinary tract infection, or sepsis); (2) all-cause hospital presentation; (3) all-cause hospital admission; and (4) all-cause emergency department visit. METHODS We regressed outcomes on demographic variables, health care use in the prior 1-year, various procedures and comorbidities related to the outcome in the prior 5 years, and prescribed medications filled in the past 120 days using modified Poisson regression to compare the risk ratio (RR) of each outcome between patients with and without ADPKD. RESULTS The median (interquartile, IQR) age was 44 (38-60 years) in the ADPKD group and 53 (42-64) in the control group. About 40% were women in both groups. The risk of ureteroscopic complications was not significantly different in patients with versus without ADPKD (8.2% vs 4.3%; adjusted RR = 1.5, 95% confidence interval [CI] = 0.7-3.2). Patients with versus without ADPKD were more likely to present to hospital after their procedure (35.6% vs. 20.0%; adjusted RR = 1.6, 95% CI = 1.2-2.2), which included a statistically significant increase in the risk of presenting to the emergency department (32.9% vs. 19.0%; adjusted RR = 1.6, 95% CI = 1.1-2.2) but not hospital admissions (10.9% vs. 5.0%; adjusted RR = 1.8, 95% CI = 0.9-3.4). LIMITATIONS The low numbers of events led to imprecision around the estimates. CONCLUSION Patients with ADPKD have a higher risk of return to the hospital within 30 days of ureteroscopy for stone disease. TRIAL REGISTRATION We did not register this study.
Collapse
Affiliation(s)
- Vinusha Kalatharan
- Department of Epidemiology and
Biostatistics, Western University, London, ON, Canada
- ICES, ON, Canada
| | - Blayne Welk
- Department of Epidemiology and
Biostatistics, Western University, London, ON, Canada
- ICES, ON, Canada
- Department of Surgery, Western
University, London, ON, Canada
| | | | | | | | - Sisira Sarma
- Department of Epidemiology and
Biostatistics, Western University, London, ON, Canada
- ICES, ON, Canada
| | - York Pei
- University Health Network, University of
Toronto, ON, Canada
| | - Amit X. Garg
- Department of Epidemiology and
Biostatistics, Western University, London, ON, Canada
- ICES, ON, Canada
- Division of Nephrology, Department of
Medicine, Western University, London, ON, Canada
| |
Collapse
|
117
|
Ha K, Nobuhara M, Wang Q, Walker RV, Qian F, Schartner C, Cao E, Delling M. The heteromeric PC-1/PC-2 polycystin complex is activated by the PC-1 N-terminus. eLife 2020; 9:60684. [PMID: 33164752 PMCID: PMC7728438 DOI: 10.7554/elife.60684] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/06/2020] [Indexed: 12/28/2022] Open
Abstract
Mutations in the polycystin proteins, PC-1 and PC-2, result in autosomal dominant polycystic kidney disease (ADPKD) and ultimately renal failure. PC-1 and PC-2 enrich on primary cilia, where they are thought to form a heteromeric ion channel complex. However, a functional understanding of the putative PC-1/PC-2 polycystin complex is lacking due to technical hurdles in reliably measuring its activity. Here we successfully reconstitute the PC-1/PC-2 complex in the plasma membrane of mammalian cells and show that it functions as an outwardly rectifying channel. Using both reconstituted and ciliary polycystin channels, we further show that a soluble fragment generated from the N-terminal extracellular domain of PC-1 functions as an intrinsic agonist that is necessary and sufficient for channel activation. We thus propose that autoproteolytic cleavage of the N-terminus of PC-1, a hotspot for ADPKD mutations, produces a soluble ligand in vivo. These findings establish a mechanistic framework for understanding the role of PC-1/PC-2 heteromers in ADPKD and suggest new therapeutic strategies that would expand upon the limited symptomatic treatments currently available for this progressive, terminal disease. On the surface of most animal and other eukaryotic cells are small rod-like protrusions known as primary cilia. Each cilium is encased by a specialized membrane which is enriched in protein complexes that help the cell sense its local environment. Some of these complexes help transport ions in out of the cell, while others act as receptors that receive chemical signals called ligands. A unique ion channel known as the polycystin complex is able to perform both of these roles as it contains a receptor called PC-1 in addition to an ion channel called PC-2. Various mutations in the genes that code for PC-1 and PC-2 can result in autosomal dominant polycystic kidney disease (ADPKD), which is the most common monogenetic disease in humans. However, due to the small size of primary cilia – which are less than a thousandth of a millimeter thick – little is known about how polycystin complexes are regulated and how mutations lead to ADPKD. To overcome this barrier, Ha et al. modified kidney cells grown in the lab so that PC-1 and PC-2 form a working channel in the plasma membrane which surrounds the entire cell. As the body of a cell is around 10,000 times bigger than the cilium, this allowed the movement of ions across the polycystin complex to be studied using conventional techniques. Experiments using this newly developed assay revealed that a region at one of the ends of the PC-1 protein, named the C-type lectin domain, is essential for stimulating polycystin complexes. Ha et al. found that this domain of PC-1 is able to cut itself from the protein complex. Further experiments showed that when fragments of PC-1, which contain the C-type lectin domain, are no longer bound to the membrane, they can activate the polycystin channels in cilia as well as the plasma membrane. This suggests that this region of PC-1 may also act as a secreted ligand that can activate other polycystin channels. Some of the genetic mutations that cause ADPKD likely disrupt the activity of the polycystin complex and reduce its ability to transport ions across the cilia membrane. Therefore, the cell assay created in this study could be used to screen for small molecules that can restore the activity of these ion channels in patients with ADPKD.
Collapse
Affiliation(s)
- Kotdaji Ha
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Mai Nobuhara
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Qinzhe Wang
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Rebecca V Walker
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, United States
| | - Feng Qian
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, United States
| | - Christoph Schartner
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| | - Erhu Cao
- Department of Biochemistry, University of Utah School of Medicine, Salt Lake City, United States
| | - Markus Delling
- Department of Physiology, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
118
|
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is the most common genetic renal disease, primarily caused by germline mutation of PKD1 or PKD2, leading to end-stage renal disease. There are few cures for ADPKD, although many researchers are trying to find a cure. The Hippo signaling pathway regulates organ growth and cell proliferation. Transcriptional coactivator with PDZ-binding motif (TAZ) is a Hippo signaling effector. In this study, we demonstrated that the PKD1–TAZ–Wnt–β-catenin–c-MYC signaling axis plays a critical role in cystogenesis. Endo IWR1 treatment, which inhibited β-catenin activity via AXIN stabilization, reduced cyst growth in an ADPKD model. Our findings provide a potential therapeutic target against ADPKD and would be important for clinical translation. Autosomal-dominant polycystic kidney disease (ADPKD) is the most common genetic renal disease, primarily caused by germline mutation of PKD1 or PKD2, leading to end-stage renal disease. The Hippo signaling pathway regulates organ growth and cell proliferation. Herein, we demonstrate the regulatory mechanism of cystogenesis in ADPKD by transcriptional coactivator with PDZ-binding motif (TAZ), a Hippo signaling effector. TAZ was highly expressed around the renal cyst-lining epithelial cells of Pkd1-deficient mice. Loss of Taz in Pkd1-deficient mice reduced cyst formation. In wild type, TAZ interacted with PKD1, which inactivated β-catenin. In contrast, in PKD1-deficient cells, TAZ interacted with AXIN1, thus increasing β-catenin activity. Interaction of TAZ with AXIN1 in PKD1-deficient cells resulted in nuclear accumulation of TAZ together with β-catenin, which up-regulated c-MYC expression. Our findings suggest that the PKD1–TAZ–Wnt–β-catenin–c-MYC signaling axis plays a critical role in cystogenesis and might be a potential therapeutic target against ADPKD.
Collapse
|
119
|
Cordido A, Cernadas E, Fernández-Delgado M, García-González MA. CystAnalyser: A new software tool for the automatic detection and quantification of cysts in Polycystic Kidney and Liver Disease, and other cystic disorders. PLoS Comput Biol 2020; 16:e1008337. [PMID: 33090995 PMCID: PMC7608985 DOI: 10.1371/journal.pcbi.1008337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 11/03/2020] [Accepted: 09/13/2020] [Indexed: 11/29/2022] Open
Abstract
The Polycystic Kidney Disease (PKD) is characterized by progressive renal cyst development and other extrarenal manifestation including Polycystic Liver Disease (PLD). Phenotypical characterization of animal models mimicking human diseases are commonly used, in order to, study new molecular mechanisms and identify new therapeutic approaches. The main biomarker of disease progression is total volume of kidney and liver in both human and mouse, which correlates with organ function. For this reason, the estimation of the number and area of the tissue occupied by cysts, is critical for the understanding of physiological mechanisms underlying the disease. In this regard, cystic index is a robust parameter commonly used to quantify the severity of the disease. To date, the vast majority of biomedical researchers use ImageJ as a software tool to estimate the cystic index by quantifying the cystic areas of histological images after thresholding. This tool has imitations of being inaccurate, largely due to incorrectly identifying non-cystic regions. We have developed a new software, named CystAnalyser (register by Universidade de Santiago de Compostela–USC, and Fundación Investigación Sanitaria de Santiago—FIDIS), that combines automatic image processing with a graphical user friendly interface that allows investigators to oversee and easily correct the image processing before quantification. CystAnalyser was able to generate a cystic profile including cystic index, number of cysts and cyst size. In order to test the CystAnalyser software, 795 cystic kidney, and liver histological images were analyzed. Using CystAnalyser there were no differences calculating cystic index automatically versus user input, except in specific circumstances where it was necessary for the user to distinguish between mildly cystic from non-cystic regions. The sensitivity and specificity of the number of cysts detected by the automatic quantification depends on the type of organ and cystic severity, with values 76.84–78.59% and 76.96–89.66% for the kidney and 87.29–93.80% and 63.42–86.07% for the liver. CystAnalyser, in addition, provides a new tool for estimating the number of cysts and a more specific measure of the cystic index than ImageJ. This study proposes CystAnalyser is a new robust and freely downloadable software tool for analyzing the severity of disease by quantifying histological images of cystic organs for routine biomedical research. CystAnalyser can be downloaded from https://citius.usc.es/transferencia/software/cystanalyser (for Windows and Linux) for research purposes. This work suggests CystAnalyser is the most reliable software tool currently available for the assessment of cystic pathologies including Polycystic Kidney Disease (PKD) and Polycystic Liver Disease (PLD). CystAnalyser combines automatic cyst recognition with a friendly graphical user interface, allowing user input prior to histological image quantification. CystAnalyser responds to the need to obtain reliable measurements of the universal biomarker for PKD and PLD disease progression, the Cystic index (area of cysts within the total area of tissue). This software tool is also able to calculate the number and size of cysts from the histological images. In summary, our results show that CystAnalyser overcomes the precision issues detected using the most commonly used software to date (ImageJ) for Cystic index quantification, offering users a reliable tool to easily characterize the phenotype and the pathophysiology of PKD and PLD in pre-clinical studies using animal models.
Collapse
Affiliation(s)
- Adrián Cordido
- Grupo de Xenética e Bioloxía do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxía (No. 11), Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain.,Grupo de Medicina Xenómica, Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain.,RedInRen RETIC, ISCIII, Spain
| | - Eva Cernadas
- Centro Singular de Investigación en Tecnoloxías Intelixentes da USC (CiTIUS) Universidade de Santiago de Compostela, Rúa Xenaro de la Fuente Domínguez, Santiago de Compostela, Spain
| | - Manuel Fernández-Delgado
- Centro Singular de Investigación en Tecnoloxías Intelixentes da USC (CiTIUS) Universidade de Santiago de Compostela, Rúa Xenaro de la Fuente Domínguez, Santiago de Compostela, Spain
| | - Miguel A García-González
- Grupo de Xenética e Bioloxía do Desenvolvemento das Enfermidades Renais, Laboratorio de Nefroloxía (No. 11), Instituto de Investigación Sanitaria de Santiago (IDIS), Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain.,Grupo de Medicina Xenómica, Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain.,RedInRen RETIC, ISCIII, Spain.,Fundación Pública Galega de Medicina Xenómica-SERGAS, Complexo Hospitalario de Santiago de Compostela (CHUS), Santiago de Compostela, Spain
| |
Collapse
|
120
|
McConnachie DJ, Stow JL, Mallett AJ. Ciliopathies and the Kidney: A Review. Am J Kidney Dis 2020; 77:410-419. [PMID: 33039432 DOI: 10.1053/j.ajkd.2020.08.012] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022]
Abstract
Primary cilia are specialized sensory organelles that protrude from the apical surface of most cell types. During the past 2 decades, they have been found to play important roles in tissue development and signal transduction, with mutations in ciliary-associated proteins resulting in a group of diseases collectively known as ciliopathies. Many of these mutations manifest as renal ciliopathies, characterized by kidney dysfunction resulting from aberrant cilia or ciliary functions. This group of overlapping and genetically heterogeneous diseases includes polycystic kidney disease, nephronophthisis, and Bardet-Biedl syndrome as the main focus of this review. Renal ciliopathies are characterized by the presence of kidney cysts that develop due to uncontrolled epithelial cell proliferation, growth, and polarity, downstream of dysregulated ciliary-dependent signaling. Due to cystic-associated kidney injury and systemic inflammation, cases result in kidney failure requiring dialysis and transplantation. Of the handful of pharmacologic treatments available, none are curative. It is important to determine the molecular mechanisms that underlie the involvement of the primary cilium in cyst initiation, expansion, and progression for the development of novel and efficacious treatments. This review updates research progress in defining key genes and molecules central to ciliogenesis and renal ciliopathies.
Collapse
Affiliation(s)
- Dominique J McConnachie
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation Disease and Research, The University of Queensland, Brisbane, QLD, Australia
| | - Jennifer L Stow
- Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia
| | - Andrew J Mallett
- Institute for Molecular Bioscience (IMB) and IMB Centre for Inflammation Disease and Research, The University of Queensland, Brisbane, QLD, Australia; Kidney Health Service, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia; KidGen Collaborative, Australian Genomics Health Alliance, Melbourne, VIC, Australia.
| |
Collapse
|
121
|
Peintner L, Venkatraman A, Waeldin A, Hofherr A, Busch T, Voronov A, Viau A, Kuehn EW, Köttgen M, Borner C. Loss of PKD1/polycystin-1 impairs lysosomal activity in a CAPN (calpain)-dependent manner. Autophagy 2020; 17:2384-2400. [PMID: 32967521 DOI: 10.1080/15548627.2020.1826716] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mutations in the PKD1 gene result in autosomal dominant polycystic kidney disease (ADPKD), the most common monogenetic cause of end-stage renal disease (ESRD) in humans. Previous reports suggested that PKD1, together with PKD2/polycystin-2, may function as a receptor-cation channel complex at cilia and on intracellular membranes and participate in various signaling pathways to regulate cell survival, proliferation and macroautophagy/autophagy. However, the exact molecular function of PKD1 and PKD2 has remained enigmatic. Here we used Pkd1-deficient mouse inner medullary collecting duct cells (mIMCD3) genetically deleted for Pkd1, and tubular epithelial cells isolated from nephrons of doxycycline-inducible conditional pkd1fl/fl;Pax8rtTA;TetOCre+ knockout mice to show that the lack of Pkd1 caused diminished lysosomal acidification, LAMP degradation and reduced CTSB/cathepsin B processing and activity. This led to an impairment of autophagosomal-lysosomal fusion, a lower delivery of ubiquitinated cargo from multivesicular bodies (MVB)/exosomes to lysosomes and an enhanced secretion of unprocessed CTSB into the extracellular space. The TFEB-dependent lysosomal biogenesis pathway was however unaffected. Pkd1-deficient cells exhibited increased activity of the calcium-dependent CAPN (calpain) proteases, probably due to a higher calcium influx. Consistent with this notion CAPN inhibitors restored lysosomal function, CTSB processing/activity and autophagosomal-lysosomal fusion, and blocked CTSB secretion and LAMP degradation in pkd1 knockout cells. Our data reveal for the first time a lysosomal function of PKD1 which keeps CAPN activity in check and ensures lysosomal integrity and a correct autophagic flux.Abbreviations: acCal: acetyl-calpastatin peptide; ADPKD: autosomal dominant polycystic kidney disease; CI-1: calpain inhibitor-1; CQ: chloroquine; Dox: doxycycline; EV: extracellular vesicles; EXO: exosomes; LAMP1/2: lysosomal-associated membrane protein 1/2; LGALS1/GAL1/galectin-1: lectin, galactose binding, soluble 1; LMP: lysosomal membrane permeabilization; mIMCD3: mouse inner medullary collecting duct cells; MV: microvesicles; MVB: multivesicular bodies; PAX8: paired box 8; PKD1/polycystin-1: polycystin 1, transient receptor potential channel interacting; PKD2/polycystin-2: polycystin 2, transient receptor potential cation channel; Tet: tetracycline; TFEB: transcription factor EB; VFM: vesicle-free medium; WT: wild-type.
Collapse
Affiliation(s)
- Lukas Peintner
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Anusha Venkatraman
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Albert Ludwigs University of Freiburg, Faculty of Biology, Freiburg, Germany
| | - Astrid Waeldin
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Alexis Hofherr
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tilman Busch
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Alexander Voronov
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Amandine Viau
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - E Wolfgang Kuehn
- Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Integrative Biological Signalling Studies (CIBSS), Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Michael Köttgen
- Spemann Graduate School of Biology and Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Renal Division, Department of Medicine, Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Integrative Biological Signalling Studies (CIBSS), Albert Ludwigs University of Freiburg, Freiburg, Germany
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Faculty of Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine, Albert Ludwigs University of Freiburg, Freiburg, Germany.,Center for Biological Signalling Studies (BIOSS), Albert Ludwigs University of Freiburg, Freiburg, Germany
| |
Collapse
|
122
|
Lacquaniti A, Campo S, Casuscelli Di Tocco T, Rovito S, Bucca M, Ragusa A, Monardo P. Acute and chronic kidney disease after pediatric liver transplantation: An underestimated problem. Clin Transplant 2020; 34:e14082. [PMID: 32949054 DOI: 10.1111/ctr.14082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 08/02/2020] [Accepted: 08/15/2020] [Indexed: 12/13/2022]
Abstract
Acute and chronic kidney injuries represent critical issues after liver transplantation (LTx), but whereas renal dysfunction in adult transplant patients is well documented, little is known about its prevalence in childhood. It is a challenge to accurately evaluate renal function in patients with liver disease, due to several confounding factors. Creatinine-based equations estimating glomerular filtration rate, validated in nephropathic patients without hepatic issues, are frequently inaccurate in end-stage liver disease, underestimating the real impact of renal disease. Moreover, whereas renal issues observed within 1 year from LTx were often related to acute injuries, kidney damage observed after 5-7 years from LTx, is due to chronic, irreversible mechanisms. Most immunosuppression protocols are based on calcineurin inhibitors (CNIs) and corticosteroids, but mycophenolate mofetil or sirolimus could play significant roles, also in children. Early diagnosis and personalized treatment represent the bases of kidney disease management, in order to minimize its close relation with increased mortality. This review analyzed acute and chronic kidney damage after pediatric LTx, also discussing the impact of pre-existent renal disease. The main immunosuppressant strategies have been reviewed, highlighting their impact on kidney function. Different methods assessing renal function were reported, with the potential application of new renal biomarkers.
Collapse
Affiliation(s)
- Antonio Lacquaniti
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Susanna Campo
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Teresa Casuscelli Di Tocco
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Stefania Rovito
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Maurizio Bucca
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Antonino Ragusa
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| | - Paolo Monardo
- Department of Internal Medicine, Nephrology and Dialysis Unit, Papardo Hospital of Messina, Messina, Italy
| |
Collapse
|
123
|
Autophagy induction promotes renal cyst growth in polycystic kidney disease. EBioMedicine 2020; 60:102986. [PMID: 32949996 PMCID: PMC7501056 DOI: 10.1016/j.ebiom.2020.102986] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 08/14/2020] [Accepted: 08/19/2020] [Indexed: 12/18/2022] Open
Abstract
Background Polycystic kidney disease (PKD) involves renal cysts arising from proliferating tubular cells. Autophagy has been recently suggested as a potential therapeutic target in PKD, and mammalian target of rapamycin (mTOR) is a key negative regulator of autophagy. However, the effect of autophagy regulation on cystogenesis has not been elucidated in PKD mice. Methods Clinical validation was performed using GEO datasets and autosomal dominant polycystic kidney disease (ADPKD) patient samples. Newly established PKD and LC3 transgenic mice were used for in vivo verifications, and additional tests were performed in vitro and in vivo using multiple autophagy drugs. Findings Neither autophagy stimulation nor LC3 overexpression alleviated PKD. Furthermore, we observed the inhibitory effect of an autophagy inhibitor on cysts, indicating its possible therapeutic use in a specific group of patients with ADPKD. Interpretation Our findings provide a novel insight into the pathogenesis related to autophagy in PKD, suggesting that drugs related to autophagy regulation should be considered with caution for treating PKD. Funding Sources This work was supported by grants from the Bio & Medical Technology Development Program; the Collaborative Genome Program for Fostering New Post-Genome Industry of the NRF; the Basic Science Program.
Collapse
|
124
|
Huang Y, Jiang K, Zhang X, Chung EJ. The effect of size, charge, and peptide ligand length on kidney targeting by small, organic nanoparticles. Bioeng Transl Med 2020; 5:e10173. [PMID: 33005739 PMCID: PMC7510478 DOI: 10.1002/btm2.10173] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/19/2020] [Accepted: 07/19/2020] [Indexed: 12/13/2022] Open
Abstract
Chronic kidney disease (CKD) affects 15% of the US adult population. However, most clinically available drugs for CKD show low bioavailability to the kidneys and non-specific uptake by other organs which results in adverse side effects. Hence, a targeted, drug delivery strategy to enhance kidney drug delivery is highly desired. Recently, our group developed small, organic nanoparticles called peptide amphiphile micelles (PAM) functionalized with the zwitterionic peptide ligand, (KKEEE)3K, that passage through the glomerular filtration barrier for kidney accumulation. Despite high bioavailability to the kidneys, these micelles also accumulated in the liver to a similar extent. To further optimize the physicochemical properties and develop design rules for kidney-targeting micelles, we synthesized a library of PAMs of varying size, charge, and peptide repeats. Specifically, variations of the original (KKEEE)3K peptide including (KKEEE)2K, (KKEEE)K, (EEKKK)3E, (EEKKK)2E, (EEKKK)E, KKKKK, and EEEEE were functionalized onto nanoparticles, and peptide surface density and PEG linker molecular weight were altered. After characterization with transmission electron microscopy (TEM) and dynamic light scattering (DLS), nanoparticles were intravenously administered into wildtype mice, and biodistribution was assessed through ex vivo imaging. All micelles localized to the kidneys, but nanoparticles that are positively-charged, close to the renal filtration size cut-off, and consisted of additional zwitterionic peptide sequences generally showed higher renal accumulation. Upon immunohistochemistry, micelles were confirmed to bind to the multiligand receptor, megalin, and histological analyses showed no tissue damage. Our study provides insight into the design of micelle carriers for kidney targeting and their potential for future therapeutic application.
Collapse
Affiliation(s)
- Yi Huang
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Kairui Jiang
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Xuting Zhang
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Eun Ji Chung
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Chemical Engineering and Materials ScienceUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Medicine, Division of Nephrology and HypertensionUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Surgery, Division of Vascular Surgery and Endovascular TherapyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
125
|
Cyst growth in ADPKD is prevented by pharmacological and genetic inhibition of TMEM16A in vivo. Nat Commun 2020; 11:4320. [PMID: 32859916 PMCID: PMC7455562 DOI: 10.1038/s41467-020-18104-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
In autosomal dominant polycystic kidney disease (ADPKD) multiple bilateral renal cysts gradually enlarge, leading to a decline in renal function. Transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and TMEM16A (anoctamin 1) are known to drive cyst enlargement. Here we demonstrate that loss of Pkd1 increased expression of TMEM16A and CFTR and Cl- secretion in murine kidneys, with TMEM16A essentially contributing to cyst growth. Upregulated TMEM16A enhanced intracellular Ca2+ signaling and proliferation of Pkd1-deficient renal epithelial cells. In contrast, increase in Ca2+ signaling, cell proliferation and CFTR expression was not observed in Pkd1/Tmem16a double knockout mice. Knockout of Tmem16a or inhibition of TMEM16A in vivo by the FDA-approved drugs niclosamide and benzbromarone, as well as the TMEM16A-specific inhibitor Ani9 largely reduced cyst enlargement and abnormal cyst cell proliferation. The present data establish a therapeutic concept for the treatment of ADPKD.
Collapse
|
126
|
Aleem M, Saleem K, Zafar S, Umer A, Arshad R, Nawaz A. Determining the Frequency of Acquired Cystic Kidney Disease in End Stage Renal Disease Patients on Hemodialysis at Dialysis Centre of Tertiary Care Hospital. Cureus 2020; 12:e10046. [PMID: 32983736 PMCID: PMC7515804 DOI: 10.7759/cureus.10046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Objectives To determine the frequency of acquired cystic kidney disease (ACKD) among patients of end-stage renal disease. Methods This cross-sectional study was conducted at the University of Lahore Teaching Hospital after approval from the ethical review committee. About 150 patients with end-stage renal disease fulfilling the inclusion criteria and undergoing three hemodialysis sessions per week for six months were approached. The patients underwent ultrasonography by the same consultant radiologist and the presence of acquired polycystic kidney disease was noted in the proforma. Data was stratified for age, gender and duration of hemodialysis and the chi-square test was applied. Results The mean age of the study participants was 47.31±9.44 years and males were majority in number with 92 (61.3%). The acquired cystic kidney disease was noted in 20 (13%) participants. There was significant difference noted in different age groups as six (6.5%) patients in the 18-40 age group and 14 (24%) patients in the 40-80 age group have acquired kidney disease (p-value=0.002). No important association between ACKD, age, and gender were found. None of these patients had evidence of renal cell carcinoma, extrarenal cysts, retroperitoneal or intrarenal hemorrhage. Conclusion There was a significant correlation between acquired cystic kidney disease and the duration of hemodialysis, and the chances of the development of acquired cystic kidney disease rise progressively with increasing time on hemodialysis.
Collapse
Affiliation(s)
- Muhammad Aleem
- Medicine, Allama Iqbal Medical College/Jinnah Hospital, Lahore, PAK
| | - Khurram Saleem
- Internal Medicine, University College of Medicine, The University of Lahore Teaching Hospital, Lahore, PAK
| | - Sana Zafar
- Internal Medicine, University College of Medicine, The University of Lahore Teaching Hospital, Lahore, PAK
| | - Amina Umer
- Medicine, Allama Iqbal Medical College/Jinnah Hospital, Lahore, PAK
| | - Rabia Arshad
- Medicine, Allama Iqbal Medical College/Jinnah Hospital, Lahore, PAK
| | - Arsalan Nawaz
- Endocrinology, Diabetes and Metabolism, Services Hospital Lahore, Lahore, PAK
| |
Collapse
|
127
|
Kalatharan V, Jandoc R, Grewal G, Nash DM, Welk B, Sarma S, Pei Y, Garg AX. Efficacy and Safety of Surgical Kidney Stone Interventions in Autosomal Dominant Polycystic Kidney Disease: A Systematic Review. Can J Kidney Health Dis 2020; 7:2054358120940433. [PMID: 32754344 PMCID: PMC7378961 DOI: 10.1177/2054358120940433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Accepted: 05/20/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Reduced kidney function and distorted kidney anatomy in patients with autosomal dominant polycystic kidney disease (ADPKD) may complicate stone interventions more compared with the general population. OBJECTIVES To review studies describing the safety and efficacy of the 3 main stone interventions in adults with ADPKD: shock wave lithotripsy (SWL), ureteroscopy, and percutaneous nephrolithotomy (PCNL). DESIGN Systematic review. SETTING Any country of origin. PATIENTS Adults with ADPKD who underwent SWL, ureteroscopy, or PCNL. MEASUREMENTS Being stone free after the intervention and postoperative complications as reported by each study, which included pain, bleeding, and fever. METHODS Relevant studies published until February 2019 were identified through a comprehensive search of MEDLINE, EMBASE, Web of Science, BIOSIS PREVIEW, and CINAHL. Studies were eligible for review if they reported at least one outcome following SWL, ureteroscopy, and/or PCNL in adults with ADPKD. We then abstracted information on study characteristics, patient characteristics, intervention details, and postintervention outcomes and assessed the methodological quality of each study using a modified Downs and Black checklist. RESULTS We screened 221 citations from which we identified 24 studies that met our review criteria. We identified an additional article when manually reviewing the reference list of an included article, yielding a total of 25 studies describing 311 patients (32 SWL, 42 ureteroscopy, and 237 PCNL). The percentage of patients who were stone free after 1 session ranged from 0% to 69% after SWL, 73% to 100% after ureteroscopy, and 45% to 100% after PCNL. The percentage of patients with ADPKD that experienced at least one postoperative complication ranged from 0% to 33% for SWL, 0% to 27% for ureteroscopy, and 0% to 100% for PCNL. LIMITATIONS The number and quality of studies published to date are limited. CONCLUSIONS The efficacy and safety of stone interventions in patients with ADPKD remains uncertain, with wide-ranging estimates reported in the literature. TRIAL REGISTRATION We did not register the protocol of this systematic review.
Collapse
Affiliation(s)
- Vinusha Kalatharan
- Department of Epidemiology &
Biostatistics, Western University, London, ON, Canada
- ICES, London, ON, Canada
| | | | - Gary Grewal
- Department of Epidemiology &
Biostatistics, Western University, London, ON, Canada
| | - Danielle M. Nash
- Department of Epidemiology &
Biostatistics, Western University, London, ON, Canada
- ICES, London, ON, Canada
| | - Blayne Welk
- Department of Epidemiology &
Biostatistics, Western University, London, ON, Canada
- ICES, London, ON, Canada
| | - Sisira Sarma
- Department of Epidemiology &
Biostatistics, Western University, London, ON, Canada
- ICES, London, ON, Canada
| | - York Pei
- Division of Nephrology, University
Health Network and University of Toronto, ON, Canada
| | - Amit X. Garg
- Department of Epidemiology &
Biostatistics, Western University, London, ON, Canada
- ICES, London, ON, Canada
- Division of Nephrology, Department of
Medicine, Western University, London, ON, Canada
| |
Collapse
|
128
|
Kim BH, Kim DY, Ahn Y, Lee EJ, Park H, Park M, Park JH. Semaphorin-3C Is Upregulated in Polycystic Kidney Epithelial Cells and Inhibits Angiogenesis of Glomerular Endothelial Cells. Am J Nephrol 2020; 51:556-564. [PMID: 32610315 DOI: 10.1159/000508263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 04/25/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND Polycystic kidney disease (PKD) is a hereditary disease characterized by cyst formation in the kidneys bilaterally. It has been observed that semaphorin-3C (SEMA3C) is overexpressed in polycystic kidney epithelial cells. It is hypothesized that upregulated SEMA3C would contribute to survival of polycystic kidney epithelial cells. Furthermore, as the kidney is a highly vascularized organ, the secreted SEMA3C from PKD epithelial cells will affect glomerular endothelial cells (GECs) in a paracrine manner. METHODS To evaluate the effect of SEMA3C on renal cells, siSEMA3C-treated PKD epithelial cells were used for further analysis, and GECs were exposed to recombinant SEMA3C (rSEMA3C). Also, co-culture and treatment of conditioned media were employed to confirm whether PKD epithelial cells could influence on GECs via SEMA3C secretion. RESULTS SEMA3C knockdown reduced proliferation of PKD epithelial cells. In case of GECs, exposure to rSEMA3C decreased angiogenesis, which resulted from suppressed migratory ability not cell proliferation. CONCLUSIONS This study indicates that SEMA3C is the aggravating factor in PKD. Thus, it is proposed that targeting SEMA3C can be effective to mitigate PKD.
Collapse
Affiliation(s)
- Bo Hye Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Do Yeon Kim
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Yejin Ahn
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Eun Ji Lee
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Hyunjoo Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Meeyoung Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea
| | - Jong Hoon Park
- Department of Biological Sciences, Sookmyung Women's University, Seoul, Republic of Korea,
| |
Collapse
|
129
|
Reciprocal Regulation between Primary Cilia and mTORC1. Genes (Basel) 2020; 11:genes11060711. [PMID: 32604881 PMCID: PMC7349257 DOI: 10.3390/genes11060711] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/22/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
In quiescent cells, primary cilia function as a mechanosensor that converts mechanic signals into chemical activities. This unique organelle plays a critical role in restricting mechanistic target of rapamycin complex 1 (mTORC1) signaling, which is essential for quiescent cells to maintain their quiescence. Multiple mechanisms have been identified that mediate the inhibitory effect of primary cilia on mTORC1 signaling. These mechanisms depend on several tumor suppressor proteins localized within the ciliary compartment, including liver kinase B1 (LKB1), AMP-activated protein kinase (AMPK), polycystin-1, and polycystin-2. Conversely, changes in mTORC1 activity are able to affect ciliogenesis and stability indirectly through autophagy. In this review, we summarize recent advances in our understanding of the reciprocal regulation of mTORC1 and primary cilia.
Collapse
|
130
|
Combined Preimplantation Genetic Testing for Autosomal Dominant Polycystic Kidney Disease: Consequences for Embryos Available for Transfer. Genes (Basel) 2020; 11:genes11060692. [PMID: 32599795 PMCID: PMC7349812 DOI: 10.3390/genes11060692] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/18/2020] [Accepted: 06/22/2020] [Indexed: 12/16/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disease and presents with genetic and clinical heterogeneity. ADPKD can also manifest extra-renally, and seminal cysts have been associated with male infertility in some cases. ADPKD-linked male infertility, along with female age, have been proposed as factors that may influence the clinical outcomes of preimplantation genetic testing (PGT) for monogenic disorders (PGT-M). Large PGT for aneuploidy assessment (PGT-A) studies link embryo aneuploidy to increasing female age; other studies suggest that embryo aneuploidy is also linked to severe male-factor infertility. We aimed to assess the number of aneuploid embryos and the number of cycles with transferable embryos in ADPKD patients after combined-PGT. The combined-PGT protocol, involving PGT-M by PCR and PGT-A by next-generation sequencing, was performed in single trophectoderm biopsies from 289 embryos in 83 PGT cycles. Transferable embryos were obtained in 69.9% of cycles. The number of aneuploid embryos and cycles with transferable embryos did not differ when the male or female had the ADPKD mutation. However, a significantly higher proportion of aneuploid embryos was found in the advanced maternal age (AMA) group, but not in the male factor (MF) group, when compared to non-AMA and non-MF groups, respectively. Additionally, no significant differences in the percentage of cycles with transferable embryos were found in any of the groups. Our results indicate that AMA couples among ADPKD patients have an increased risk of aneuploid embryos, but ADPKD-linked male infertility does not promote an increased aneuploidy rate.
Collapse
|
131
|
Millet-Boureima C, Selber-Hnatiw S, Gamberi C. Drug discovery and chemical probing in Drosophila. Genome 2020; 64:147-159. [PMID: 32551911 DOI: 10.1139/gen-2020-0037] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Flies are increasingly utilized in drug discovery and chemical probing in vivo, which are novel technologies complementary to genetic probing in fundamental biological studies. Excellent genetic conservation, small size, short generation time, and over one hundred years of genetics make Drosophila an attractive model for rapid assay readout and use of analytical amounts of compound, enabling the experimental iterations needed in early drug development at a fraction of time and costs. Here, we describe an effective drug-testing pipeline using adult flies that can be easily implemented to study several disease models and different genotypes to discover novel molecular insight, probes, quality lead compounds, and develop novel prototype drugs.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Susannah Selber-Hnatiw
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| | - Chiara Gamberi
- Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada.,Biology Department, Concordia University, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
132
|
Molinari E, Sayer JA. Disease Modeling To Understand the Pathomechanisms of Human Genetic Kidney Disorders. Clin J Am Soc Nephrol 2020; 15:855-872. [PMID: 32139361 PMCID: PMC7274277 DOI: 10.2215/cjn.08890719] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The class of human genetic kidney diseases is extremely broad and heterogeneous. Accordingly, the range of associated disease phenotypes is highly variable. Many children and adults affected by inherited kidney disease will progress to ESKD at some point in life. Extensive research has been performed on various different disease models to investigate the underlying causes of genetic kidney disease and to identify disease mechanisms that are amenable to therapy. We review some of the research highlights that, by modeling inherited kidney disease, contributed to a better understanding of the underlying pathomechanisms, leading to the identification of novel genetic causes, new therapeutic targets, and to the development of new treatments. We also discuss how the implementation of more efficient genome-editing techniques and tissue-culture methods for kidney research is providing us with personalized models for a precision-medicine approach that takes into account the specificities of the patient and the underlying disease. We focus on the most common model systems used in kidney research and discuss how, according to their specific features, they can differentially contribute to biomedical research. Unfortunately, no definitive treatment exists for most inherited kidney disorders, warranting further exploitation of the existing disease models, as well as the implementation of novel, complex, human patient-specific models to deliver research breakthroughs.
Collapse
Affiliation(s)
- Elisa Molinari
- Faculty of Medical Sciences, Translational and Clinical Research Institute, International Centre for Life, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John A. Sayer
- Faculty of Medical Sciences, Translational and Clinical Research Institute, International Centre for Life, Newcastle University, Newcastle upon Tyne, United Kingdom
- Renal Services, Newcastle Upon Tyne Hospitals National Health Service Trust, Newcastle upon Tyne, United Kingdom
- National Institute for Health Research Newcastle Biomedical Research Centre, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
133
|
Ganoderic acid A is the effective ingredient of Ganoderma triterpenes in retarding renal cyst development in polycystic kidney disease. Acta Pharmacol Sin 2020; 41:782-790. [PMID: 31911637 PMCID: PMC7468358 DOI: 10.1038/s41401-019-0329-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is one of the most common life-threatening monogenetic diseases characterized by progressive enlargement of fluid-filled renal cysts. Our previous study has shown that Ganoderma triterpenes (GT) retards PKD renal cyst development. In the present study we identified the effective ingredient of GT in suppression of kidney cyst development. Using an in vitro MDCK cystogenesis model, we identified ganoderic acid A (GA-A) as the most promising candidate among the 12 ganoderic acid (GA) monomers. We further showed that GA-A (6.25−100 μM) significantly inhibited cyst growth in MDCK cyst model and embryonic kidney cyst model in vitro, and the inhibitory effect was reversible. In kidney-specific Pkd1 knockout (kPKD) mice displaying severe cystic kidney disease, administration of GA-A (50 mg· kg−1 ·d−1, sc) significantly attenuated renal cyst development. In both MDCK cells and kidney of kPKD mice, we revealed that GA-A dose-dependently downregulated the Ras/MAPK signaling pathway. The expression of proliferating cell nuclear antigen (PCNA) was also suppressed, suggesting a possible effect of GA-A on cell proliferation. These experimental data suggest that GA-A may be the main ingredient of GT as a potential therapeutic reagent for treating ADPKD.
Collapse
|
134
|
Abdelwahed M, Hilbert P, Ahmed A, Dey M, Kamoun H, Ammar-Keskes L, Belguith N. Detection of a novel mutation in a Tunisian child with polycystic kidney disease. IUBMB Life 2020; 72:1799-1806. [PMID: 32472977 DOI: 10.1002/iub.2309] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/08/2020] [Accepted: 05/11/2020] [Indexed: 11/06/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common monogenic disease that has an adverse impact on the patients' health and quality of life. ADPKD is usually known as "adult-type disease," but rare cases have been reported in pediatric patients. We present here a 2-year-old Tunisian girl with renal cyst formation and her mother with adult onset ADPKD. Disease-causing mutation has been searched in PKD1 and PKD2 using Long-Range and PCR followed by sequencing. Molecular sequencing displayed us to identify a novel likely pathogenic mutation (c.696 T > G; p.C232W, exon 5) in PKD1. The identified PKD1 mutation is inherited and unreported variant, which can alter the formation of intramolecular disulfide bonds essential for polycystin-1 function. We report here the first mutational study in pediatric patient with ADPKD in Tunisia.
Collapse
Affiliation(s)
- Mayssa Abdelwahed
- Laboratory of Human Molecular Genetics, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Pascale Hilbert
- Center of Human Genetics, Institute of Pathology and Genetics, Gosselies, Belgium
| | - Asma Ahmed
- Nephrology and Hemodialyse Department, Mohamed Ben Sassi Hospital, Gabes, Tunisia
| | - Mouna Dey
- Nephrology and Hemodialyse Department, Mohamed Ben Sassi Hospital, Gabes, Tunisia
| | - Hassen Kamoun
- Medical Genetics Department, HediChaker Hospital, Sfax, Tunisia
| | - Leila Ammar-Keskes
- Laboratory of Human Molecular Genetics, Faculty of Medicine, University of Sfax, Sfax, Tunisia
| | - Neïla Belguith
- Laboratory of Human Molecular Genetics, Faculty of Medicine, University of Sfax, Sfax, Tunisia.,Medical Genetics Department, HediChaker Hospital, Sfax, Tunisia
| |
Collapse
|
135
|
Wilson EM, Choi J, Torres VE, Somlo S, Besse W. Large Deletions in GANAB and SEC63 Explain 2 Cases of Polycystic Kidney and Liver Disease. Kidney Int Rep 2020; 5:727-731. [PMID: 32405593 PMCID: PMC7210741 DOI: 10.1016/j.ekir.2020.01.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/09/2020] [Accepted: 01/20/2020] [Indexed: 11/29/2022] Open
Affiliation(s)
- Elena M Wilson
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jungmin Choi
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Stefan Somlo
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, Connecticut, USA.,Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Whitney Besse
- Department of Internal Medicine, Section of Nephrology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
136
|
Cadamuro M, Girardi N, Gores GJ, Strazzabosco M, Fabris L. The Emerging Role of Macrophages in Chronic Cholangiopathies Featuring Biliary Fibrosis: An Attractive Therapeutic Target for Orphan Diseases. Front Med (Lausanne) 2020; 7:115. [PMID: 32373615 PMCID: PMC7186419 DOI: 10.3389/fmed.2020.00115] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 03/12/2020] [Indexed: 12/16/2022] Open
Abstract
Cholangiopathies are a heterogeneous group of chronic liver diseases caused by different types of injury targeting the biliary epithelium, such as genetic defects and immune-mediated attacks. Notably, most cholangiopathies are orphan, thereby representing one of the major gaps in knowledge of the modern hepatology. A typical hallmark of disease progression in cholangiopathies is portal scarring, and thus development of effective therapeutic approaches would aim to hinder cellular and molecular mechanisms underpinning biliary fibrogenesis. Recent lines of evidence indicate that macrophages, rather than more conventional cell effectors of liver fibrosis such as hepatic stellate cells and portal fibroblasts, are actively involved in the earliest stages of biliary fibrogenesis by exchanging a multitude of cues with cholangiocytes, which promote their recruitment from the circulating compartment owing to a senescent or an immature epithelial phenotype. Two cholangiopathies, namely primary sclerosing cholangitis and congenital hepatic fibrosis, are paradigmatic of this mechanism. This review summarizes current understandings of the cytokine and extracellular vesicles-mediated communications between cholangiocytes and macrophages typically occurring in the two cholangiopathies to unveil potential novel targets for the treatment of biliary fibrosis.
Collapse
Affiliation(s)
| | - Noemi Girardi
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology and the Mayo Clinic Center for Cell Signaling in Gastroenterology, Mayo Clinic, Rochester, NY, United States
| | - Mario Strazzabosco
- Liver Center, Department of Medicine, Yale University, New Haven, CT, United States
| | - Luca Fabris
- Department of Molecular Medicine, University of Padua, Padua, Italy.,Liver Center, Department of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
137
|
Liu W, Wang Y. Large-scale analysis of zebrafish (Danio rerio) transcriptomes identifies functional modules associated with phenotypes. Mar Genomics 2020; 53:100770. [PMID: 32276847 DOI: 10.1016/j.margen.2020.100770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 03/26/2020] [Accepted: 03/28/2020] [Indexed: 11/28/2022]
Abstract
Zebrafish (Danio rerio) is an excellent model for biomedicine research due to its genetic accessibility and optical transparency. A large number of microarray based transcriptomes of zebrafish have been profiled in various cell types, tissues, development stages, toxicological exposures and other conditions. However, there is still no easy-to-use web tool to explore those precious data. We downloaded 1434 microarray data from National Center for Biotechnology Information Gene Expression Omnibus (NCBI GEO), constructed weighted gene co-expression network, and identified 50 modules of co-expressed genes that correspond to different cell types, tissues, development stages, and other experimental conditions. These modules were associated with experiments/traits, and may serve signature modules for phenotypes. Hub genes were screened by intra-modular connectivity. Higher-order module networks analysis suggested that nucleus and cell cycle modules are densely connected. Module-based gene function identification may help to discover novel gene function. Our web tool provides a new resource for gene function study in zebrafish (http://bioinformatics.fafu.edu.cn/zebrafish/).
Collapse
Affiliation(s)
- Wei Liu
- School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| | - Yanan Wang
- School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| |
Collapse
|
138
|
Cassina L, Chiaravalli M, Boletta A. Increased mitochondrial fragmentation in polycystic kidney disease acts as a modifier of disease progression. FASEB J 2020; 34:6493-6507. [PMID: 32239723 DOI: 10.1096/fj.201901739rr] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 02/17/2020] [Accepted: 03/06/2020] [Indexed: 12/23/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a common monogenic disorder, characterized by bilateral renal cyst formation. Multiple pathways are de-regulated in cystic epithelia offering good opportunities for therapy. Others and we have previously reported that metabolic reprogramming, including alterations of the TCA cycle, are prominent features of ADPKD. Several lines of evidence suggest that mitochondrial impairment might be responsible for the metabolic alterations. Here, we performed morphologic and morphometric evaluation of mitochondria by TEM in an orthologous mouse model of PKD caused by mutations in the Pkd1 gene (Ksp-Cre;Pkd1flox/- ). Furthermore, we measured mitochondrial respiration by COX and SDH enzymatic activity in situ. We found several alterations including reduced mitochondrial mass, altered structure and fragmentation of the mitochondrial network in cystic epithelia of Ksp-Cre;Pkd1flox/- mice. At the molecular level, we found reduced expression of the pro-fusion proteins OPA1 and MFN1 and up-regulation of the pro-fission protein DRP1. Importantly, administration of Mdivi-1, which interferes with DRP1 rescuing mitochondrial fragmentation, significantly reduced kidney/body weight, cyst formation, and improved renal function in Ksp-Cre;Pkd1flox/- mice. Our data indicate that impaired mitochondrial structure and function play a role in disease progression, and that their improvement can significantly modify the course of the disease.
Collapse
Affiliation(s)
- Laura Cassina
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS-San Raffaele Scientific Institute, Milan, Italy
| | - Marco Chiaravalli
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS-San Raffaele Scientific Institute, Milan, Italy
| | - Alessandra Boletta
- Molecular Basis of Cystic Kidney Disorders Unit, Division of Genetics and Cell Biology, IRCCS-San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
139
|
Zimmerman KA, Huang J, He L, Revell DZ, Li Z, Hsu JS, Fitzgibbon WR, Hazard ES, Hardiman G, Mrug M, Bell PD, Yoder BK, Saigusa T. Interferon Regulatory Factor-5 in Resident Macrophage Promotes Polycystic Kidney Disease. ACTA ACUST UNITED AC 2020; 1:179-190. [PMID: 33490963 DOI: 10.34067/kid.0001052019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Autosomal dominant polycystic kidney disease is caused by genetic mutations in PKD1 or PKD2. Macrophages and their associated inflammatory cytokines promote cyst progression; however, transcription factors within macrophages that control cytokine production and cystic disease are unknown. Methods In these studies, we used conditional Pkd1 mice to test the hypothesis that macrophage-localized interferon regulatory factor-5 (IRF5), a transcription factor associated with production of cyst-promoting cytokines (TNFα, IL-6), is required for accelerated cyst progression in a unilateral nephrectomy (1K) model. Analyses of quantitative real-time PCR (qRT-PCR) and flow-cytometry data 3 weeks post nephrectomy, a time point before the onset of severe cystogenesis, indicate an accumulation of inflammatory infiltrating and resident macrophages in 1K Pkd1 mice compared with controls. qRT-PCR data from FACS cells at this time demonstrate that macrophages from 1K Pkd1 mice have increased expression of Irf5 compared with controls. To determine the importance of macrophage-localized Irf5 in cyst progression, we injected scrambled or IRF5 antisense oligonucleotide (ASO) in 1K Pkd1 mice and analyzed the effect on macrophage numbers, cytokine production, and renal cystogenesis 6 weeks post nephrectomy. Results Analyses of qRT-PCR and IRF5 ASO treatment significantly reduced macrophage numbers, Irf5 expression in resident-but not infiltrating-macrophages, and the severity of cystic disease. In addition, IRF5 ASO treatment in 1K Pkd1 mice reduced Il6 expression in resident macrophages, which was correlated with reduced STAT3 phosphorylation and downstream p-STAT3 target gene expression. Conclusions These data suggest that Irf5 promotes inflammatory cytokine production in resident macrophages resulting in accelerated cystogenesis.
Collapse
Affiliation(s)
- Kurt A Zimmerman
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jifeng Huang
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Lan He
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Dustin Z Revell
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Zhang Li
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jung-Shan Hsu
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Wayne R Fitzgibbon
- Division of Nephrology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - E Starr Hazard
- Academic Affairs Faculty and Computational Biology Resource Center, Medical University of South Carolina, Charleston, South Carolina
| | - Gary Hardiman
- School of Biological Sciences, Institute for Global Food Security, Queens University Belfast, Belfast, United Kingdom
| | - Michal Mrug
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama.,Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - P Darwin Bell
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| | - Bradley K Yoder
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Takamitsu Saigusa
- Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
140
|
TMEM16A drives renal cyst growth by augmenting Ca 2+ signaling in M1 cells. J Mol Med (Berl) 2020; 98:659-671. [PMID: 32185407 PMCID: PMC7220898 DOI: 10.1007/s00109-020-01894-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 02/27/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Abstract Polycystic kidney disease (PKD) leads to continuous decline of renal function by growth of renal cysts. Enhanced proliferation and transepithelial chloride secretion through cystic fibrosis transmembrane conductance regulator (CFTR) and Ca2+-activated TMEM16A Cl− channels is thought to cause an increase in cyst volume. Recent work shows the pro-proliferative role of the Ca2+ activated Cl− channel TMEM16A (anoctamin 1), and demonstrates the essential contribution of TMEM16A to CFTR-dependent Cl− secretion. The present data demonstrate an increase in intracellular Ca2+ ([Ca2+]i) signals and Cl− secretion by TMEM16A, in renal collecting duct principle cells from dog (MDCK) and mouse (M1) as well as primary tubular epithelial cells from PKD1−/− knockout mice. M1 organoids proliferated, increased expression of TMEM16A, and secreted Cl− upon knockdown of endogenous polycystin 1 or 2 (PKD1,2), by retroviral transfection with shPKD1 and shPKD2, respectively. Knockdown of PKD1 or PKD2 increased basal intracellular Ca2+ levels and enhanced purinergic Ca2+ release from endoplasmic reticulum. In contrast, ryanodine receptors were found not to be expressed in mouse renal epithelial cells and caffeine had no effects on [Ca2+]i. Ca2+ signals, proliferation, and Cl− secretion were largely reduced by knockdown or blockade of TMEM16A. TMEM16A may be therefore important for enhanced Ca2+ release from IP3-sensitive Ca2+ stores in polycystic kidney disease. Key messages • ADPKD leads to continuous decline of renal function by growth of renal cysts. • Knockdown of PKD1 or PKD2 increases TMEM16A expression. • TMEM16A enhanced intracellular Ca2+ signals, Cl− secretion, and proliferation. • TMEM16A contributes to cyst growth in ADPKD. Electronic supplementary material The online version of this article (10.1007/s00109-020-01894-y) contains supplementary material, which is available to authorized users.
Collapse
|
141
|
Abstract
Mutations in the polycystins PC1 or PC2 cause autosomal dominant polycystic kidney disease (ADPKD), which is characterized by the formation of fluid-filled renal cysts that disrupt renal architecture and function, ultimately leading to kidney failure in the majority of patients. Although the genetic basis of ADPKD is now well established, the physiological function of polycystins remains obscure and a matter of intense debate. The structural determination of both the homomeric PC2 and heteromeric PC1-PC2 complexes, as well as the electrophysiological characterization of PC2 in the primary cilium of renal epithelial cells, provided new valuable insights into the mechanisms of ADPKD pathogenesis. Current findings indicate that PC2 can function independently of PC1 in the primary cilium of renal collecting duct epithelial cells to form a channel that is mainly permeant to monovalent cations and is activated by both membrane depolarization and an increase in intraciliary calcium. In addition, PC2 functions as a calcium-activated calcium release channel at the endoplasmic reticulum membrane. Structural studies indicate that the heteromeric PC1-PC2 complex comprises one PC1 and three PC2 channel subunits. Surprisingly, several positively charged residues from PC1 occlude the ionic pore of the PC1-PC2 complex, suggesting that pathogenic polycystin mutations might cause ADPKD independently of an effect on channel permeation. Emerging reports of novel structural and functional findings on polycystins will continue to elucidate the molecular basis of ADPKD.
Collapse
|
142
|
Kim H, Sung J, Kim H, Ryu H, Cho Park H, Oh YK, Lee HS, Oh KH, Ahn C. Expression and secretion of CXCL12 are enhanced in autosomal dominant polycystic kidney disease. BMB Rep 2020. [PMID: 31186083 PMCID: PMC6675246 DOI: 10.5483/bmbrep.2019.52.7.112] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD), one of the most common human monogenic diseases (frequency of 1/1000-1/400), is characterized by numerous fluid-filled renal cysts (RCs). Inactivation of the PKD1 or PKD2 gene by germline and somatic mutations is necessary for cyst formation in ADPKD. To mechanistically understand cyst formation and growth, we isolated RCs from Korean patients with ADPKD and immortalized them with human telomerase reverse transcriptase (hTERT). Three hTERT-immortalized RC cell lines were characterized as proximal epithelial cells with germline and somatic PKD1 mutations. Thus, we first established hTERT-immortalized proximal cyst cells with somatic PKD1 mutations. Through transcriptome sequencing and Gene Ontology (GO) analysis, we found that upregulated genes were related to cell division and that downregulated genes were related to cell differentiation. We wondered whether the upregulated gene for the chemokine CXCL12 is related to the mTOR signaling pathway in cyst growth in ADPKD. CXCL12 mRNA expression and secretion were increased in RC cell lines. We then examined CXCL12 levels in RC fluids from patients with ADPKD and found increased CXCL12 levels. The CXCL12 receptor CXC chemokine receptor 4 (CXCR4) was upregulated, and the mTOR signaling pathway, which is downstream of the CXCL12/CXCR4 axis, was activated in ADPKD kidney tissue. To confirm activation of the mTOR signaling pathway by CXCL12 via CXCR4, we treated the RC cell lines with recombinant CXCL12 and the CXCR4 antagonist AMD3100; CXCL12 induced the mTOR signaling pathway, but the CXCR4 antagonist AMD3100 blocked the mTOR signaling pathway. Taken together, these results suggest that enhanced CXCL12 in RC fluids activates the mTOR signaling pathway via CXCR4 in ADPKD cyst growth.
Collapse
Affiliation(s)
- Hyunho Kim
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Jinmo Sung
- Center for Medical Innovation, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Hyunsuk Kim
- Internal Medicine, Hallym University Medical Center, Chuncheon Sacred Heart Hospital, Chuncheon 24253, Korea
| | - Hyunjin Ryu
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Hayne Cho Park
- Department of Internal Medicine, Hallym University Medical Center, Kangnam Sacred Heart Hospital, Seoul 07441, Korea
| | - Yun Kyu Oh
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul 07061, Korea
| | - Hyun-Seob Lee
- Genomics Core Facility, Department of Transdisciplinary Research and Collaboration, Biomedical Research Institute, Seoul National University Hospital, Seoul 03082, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Curie Ahn
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
143
|
Cohen E, Sawyer JK, Peterson NG, Dow JAT, Fox DT. Physiology, Development, and Disease Modeling in the Drosophila Excretory System. Genetics 2020; 214:235-264. [PMID: 32029579 PMCID: PMC7017010 DOI: 10.1534/genetics.119.302289] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 11/04/2019] [Indexed: 02/06/2023] Open
Abstract
The insect excretory system contains two organ systems acting in concert: the Malpighian tubules and the hindgut perform essential roles in excretion and ionic and osmotic homeostasis. For over 350 years, these two organs have fascinated biologists as a model of organ structure and function. As part of a recent surge in interest, research on the Malpighian tubules and hindgut of Drosophila have uncovered important paradigms of organ physiology and development. Further, many human disease processes can be modeled in these organs. Here, focusing on discoveries in the past 10 years, we provide an overview of the anatomy and physiology of the Drosophila excretory system. We describe the major developmental events that build these organs during embryogenesis, remodel them during metamorphosis, and repair them following injury. Finally, we highlight the use of the Malpighian tubules and hindgut as accessible models of human disease biology. The Malpighian tubule is a particularly excellent model to study rapid fluid transport, neuroendocrine control of renal function, and modeling of numerous human renal conditions such as kidney stones, while the hindgut provides an outstanding model for processes such as the role of cell chirality in development, nonstem cell-based injury repair, cancer-promoting processes, and communication between the intestine and nervous system.
Collapse
Affiliation(s)
| | - Jessica K Sawyer
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, and
| | | | - Julian A T Dow
- Institute of Molecular, Cell, and Systems Biology, University of Glasgow, G12 8QQ, United Kingdom
| | - Donald T Fox
- Department of Cell Biology and
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina 27710, and
| |
Collapse
|
144
|
Akarkach A, Burgmaier K, Sander A, Hooman N, Sever L, Cano F, Zambrano P, Bilge I, Flynn JT, Yavascan O, Vallés PG, Munarriz RL, Patel HP, Serdaroglu E, Koch VH, Suarez ADC, Galanti M, Celedon CG, Rébori A, Kari JA, Wong CJ, Elenberg E, Rojas LF, Warady BA, Liebau MC, Schaefer F. Maintenance Peritoneal Dialysis in Children With Autosomal Recessive Polycystic Kidney Disease: A Comparative Cohort Study of the International Pediatric Peritoneal Dialysis Network Registry. Am J Kidney Dis 2020; 75:460-464. [PMID: 31983502 DOI: 10.1053/j.ajkd.2019.10.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 10/06/2019] [Indexed: 01/18/2023]
Affiliation(s)
- Abdelaziz Akarkach
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, Cologne, Germany
| | - Kathrin Burgmaier
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, Cologne, Germany
| | - Anja Sander
- Institute of Medical Biometry and Informatics, University of Heidelberg, Heidelberg, Germany
| | - Nakysa Hooman
- Ali-Asghar Clinical Research Development Center, Iran University of Medical Sciences, Tehran, Iran
| | - Lale Sever
- Pediatric Nephrology, Cerrahpaşa Faculty of Medicine, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Francisco Cano
- Division of Pediatrics, Luis Calvo Mackenna Children's Hospital, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | - Ilmay Bilge
- Department of Pediatric Nephrology, Istanbul University Medical Faculty, Istanbul, Turkey
| | - Joseph T Flynn
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA; Division of Nephrology, Seattle Children's Hospital, Seattle, WA
| | - Onder Yavascan
- Department of Pediatric Nephrology, Tepecik Teaching and Research Hospital, Izmir, Turkey
| | | | | | - Hiren P Patel
- Department of Pediatrics, Ohio State University College of Medicine and Nationwide Children's Hospital, Columbus, OH
| | - Erkin Serdaroglu
- Department of Pediatric Nephrology, Dr. Behçet Uz Children's Hospital, Izmir, Turkey
| | - Vera H Koch
- Pediatric Nephrology Unit, Instituto da Criança Hospital das Clinicas, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Angela Del Carmen Suarez
- Department of Nephrology, Hospital de Niños Sor María Ludovica La Plata, Buenos Aires, Argentina
| | - Monica Galanti
- Pediatric Nephrology, Roberto del Río Hospital, Santiago, Chile
| | | | - Anabella Rébori
- Pediatric Dialysis Unit, Senniad, Hospital Evangelico, Montevideo, Uruguay
| | - Jameela A Kari
- Pediatric Nephrology Center of Excellence and Pediatric Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Cynthia J Wong
- Division of Nephrology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA
| | | | - Luisa F Rojas
- Baxter Servicio al Cliente Colombia, Medellin-Antioquia, Colombia
| | - Bradley A Warady
- Division of Nephrology, Children's Mercy Kansas City, University of Missouri-Kansas City School of Medicine, Kansas City, MO
| | - Max C Liebau
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Pediatrics, Cologne, Germany; Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany.
| | - Franz Schaefer
- Division of Pediatric Nephrology, Center for Pediatrics and Adolescent Medicine, University Hospital of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
145
|
Kalatharan V, McArthur E, Nash DM, Welk B, Sarma S, Garg AX, Pei Y. Diagnostic accuracy of administrative codes for autosomal dominant polycystic kidney disease in clinic patients with cystic kidney disease. Clin Kidney J 2020; 14:612-616. [PMID: 33623686 PMCID: PMC7886566 DOI: 10.1093/ckj/sfz184] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Accepted: 11/08/2019] [Indexed: 01/03/2023] Open
Abstract
Background The ability to identify patients with autosomal dominant polycystic kidney disease (ADPKD) and distinguish them from patients with similar conditions in healthcare administrative databases is uncertain. We aimed to measure the sensitivity and specificity of different ADPKD administrative coding algorithms in a clinic population with non-ADPKD and ADPKD kidney cystic disease. Methods We used a dataset of all patients who attended a hereditary kidney disease clinic in Toronto, Ontario, Canada between 1 January 2010 and 23 December 2014. This dataset included patients who met our reference standard definition of ADPKD or other cystic kidney disease. We linked this dataset to healthcare databases in Ontario. We developed eight algorithms to identify ADPKD using the International Classification of Diseases, 10th Revision (ICD-10) codes and provincial diagnostic billing codes. A patient was considered algorithm positive if any one of the codes in the algorithm appeared at least once between 1 April 2002 and 31 March 2015. Results The ICD-10 coding algorithm had a sensitivity of 33.7% [95% confidence interval (CI) 30.0–37.7] and a specificity of 86.2% (95% CI 75.7–92.5) for the identification of ADPKD. The provincial diagnostic billing code had a sensitivity of 91.1% (95% CI 88.5–93.1) and a specificity of 10.8% (95% CI 5.3–20.6). Conclusions ICD-10 coding may be useful to identify patients with a high chance of having ADPKD but fail to identify many patients with ADPKD. Provincial diagnosis billing codes identified most patients with ADPKD and also with other types of cystic kidney disease.
Collapse
Affiliation(s)
- Vinusha Kalatharan
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada
| | | | | | - Blayne Welk
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada.,ICES, London, Ontario, Canada
| | - Sisira Sarma
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada.,ICES, London, Ontario, Canada
| | - Amit X Garg
- Department of Epidemiology and Biostatistics, Western University, London, Ontario, Canada.,ICES, London, Ontario, Canada.,Division of Nephrology, Department of Medicine, Western University, London, Ontario, Canada
| | - York Pei
- University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
146
|
von Kodolitsch Y, Demolder A, Girdauskas E, Kaemmerer H, Kornhuber K, Muino Mosquera L, Morris S, Neptune E, Pyeritz R, Rand-Hendriksen S, Rahman A, Riise N, Robert L, Staufenbiel I, Szöcs K, Vanem TT, Linke SJ, Vogler M, Yetman A, De Backer J. Features of Marfan syndrome not listed in the Ghent nosology – the dark side of the disease. Expert Rev Cardiovasc Ther 2020; 17:883-915. [DOI: 10.1080/14779072.2019.1704625] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Yskert von Kodolitsch
- German Aorta Center Hamburg at University Hospital Hamburg Eppendorf University Heart Centre, Clinics for Cardiology and Heart Surgery, VASCERN HTAD European Reference Centre
| | - Anthony Demolder
- Center for Medical Genetics and Department of Cardiology, Ghent University Hospital, VASCERN HTAD European Reference Centre, Ghent, Belgium
| | - Evaldas Girdauskas
- German Aorta Center Hamburg at University Hospital Hamburg Eppendorf University Heart Centre, Clinics for Cardiology and Heart Surgery, VASCERN HTAD European Reference Centre
| | - Harald Kaemmerer
- Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich of the Free State of Bavaria, Munich
| | - Katharina Kornhuber
- Department of Pediatric Cardiology and Congenital Heart Disease, German Heart Centre Munich of the Free State of Bavaria, Munich
| | - Laura Muino Mosquera
- Department of Pediatric Cardiology and Center for Medical Genetics, Ghent University Hospital, Ghent, Belgium
| | - Shaine Morris
- Department of Pediatrics-Cardiology, Texas Children’s Hospital/Baylor College of Medicine, Houston, TX, USA
| | - Enid Neptune
- Division of Pulmonary and Critical Care Medicine and Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Reed Pyeritz
- Departments of Medicine and Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Svend Rand-Hendriksen
- TRS, National Resource Centre for Rare Disorders, Sunnaas Rehabilitation Hospital, Nesoddtangen, Norway
| | - Alexander Rahman
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, Hannover
| | - Nina Riise
- TRS, National Resource Centre for Rare Disorders, Sunnaas Rehabilitation Hospital, Nesoddtangen, Norway
| | - Leema Robert
- Department of Clinical Genetics, Guy’s and St Thomas’ NHS Foundation Trust, London, UK
| | - Ingmar Staufenbiel
- Department of Conservative Dentistry, Periodontology and Preventive Dentistry, Hannover Medical School, Hannover
| | - Katalin Szöcs
- German Aorta Center Hamburg at University Hospital Hamburg Eppendorf University Heart Centre, Clinics for Cardiology and Heart Surgery, VASCERN HTAD European Reference Centre
| | - Thy Thy Vanem
- TRS, National Resource Centre for Rare Disorders, Sunnaas Rehabilitation Hospital, Nesoddtangen, Norway
- Department of Physical Medicine and Rehabilitation, Oslo University Hospital, Oslo, Norway
| | - Stephan J. Linke
- Clinic of Ophthalmology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Ophthalmological practice at the University Clinic Hamburg-Eppendorf, zentrumsehstärke, Hamburg, Germany
| | - Marina Vogler
- German Marfan Association, Marfan Hilfe Deutschland e.V, Eutin, Germany
| | - Anji Yetman
- Vascular Medicine, Children’s Hospital and Medical Center, Omaha, USA
| | - Julie De Backer
- Center for Medical Genetics and Department of Cardiology, Ghent University Hospital, VASCERN HTAD European Reference Centre, Ghent, Belgium
| |
Collapse
|
147
|
Claudin-7b and Claudin-h are required for controlling cilia morphogenesis in the zebrafish kidney. Mech Dev 2019; 161:103595. [PMID: 31887432 DOI: 10.1016/j.mod.2019.103595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 12/08/2019] [Accepted: 12/23/2019] [Indexed: 11/22/2022]
Abstract
Claudins are a family of proteins which are the most important components of the tight junctions. The location of Claudins on the renal tubule epithelial determines its paracellular transport characteristics, but whether Claudins have other functions in kidneys remains still unclear. Here, we showed that the transcripts encoding two Claudin family proteins, claudin-7b (cldn-7b) and claudin-h (cldn-h), were expressed in the transporting cells in the zebrafish pronephros. By knocking down of cldn-7b and cldn-h in zebrafish, we showed that these claudins morphants exhibited cystic kidneys accompanied with body curvature. Further analysis showed that down regulation of cldn-7b or cldn-h led to multiple defects in apico-basolateral polarity, cilia morphology and ciliary function in kidney. Moreover, the ciliary defect was confirmed by depletion of Cldn-7b or Cldn-h using CRISPR/Cas9 system. We also showed that both cldn-7b and cldn-h were genetically interacted with a well-known ciliary gene, arl13b. Deletion of arl13b led to curly cilia in the pronephros that phenocopied with cldn-7b and cldn-h morphants. Taken together, our data suggested that the tight junction protein, Cldn-7b and Cldn-h, regulate kidney development and function by affecting cilia morphology.
Collapse
|
148
|
Raina R, Chauvin A, Chakraborty R, Nair N, Shah H, Krishnappa V, Kusumi K. The Role of Endothelin and Endothelin Antagonists in Chronic Kidney Disease. KIDNEY DISEASES 2019; 6:22-34. [PMID: 32021871 DOI: 10.1159/000504623] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/30/2019] [Indexed: 12/21/2022]
Abstract
Background Endothelins (ET) are a family of peptides that act as potent vasoconstrictors and pro-fibrotic growth factors. ET-1 is integral to renal and cardiovascular pathophysiology and exerts effects via autocrine, paracrine and endocrine signaling pathways tied to regulation of aldosterone, catecholamines, and angiotensin. In the kidney, ET-1 is critical to maintaining renal perfusion and controls glomerular arteriole tone and hemodynamics. It is hypothesized that ET-1 influences the progression of chronic kidney disease (CKD), and the objective of this review is to discuss the pathophysiology, and role of ET and endothelin receptor antagonists (ERAs) in CKD. Summary The use of ERAs in hypertensive nephropathy has the potential to decrease proteinuria, and in diabetic nephropathy has the potential to restore glycocalyx thickness, also decreasing proteinuria. Focal segmental glomerular sclerosis has no specific Food and Drug Administration-approved therapy currently, however, ERAs show promise in decreasing proteinuria and slowing tissue damage. ET-1 is a potential biomarker for autosomal dominant polycystic kidney disease progression and so it is thought that ERAs may be of some therapeutic benefit. Key Messages Multiple studies have shown the utility of ERAs in CKD. These agents have shown to reduce blood pressure, proteinuria, and arterial stiffness. However, more clinical trials are needed, and the results of active or recently concluded studies are eagerly awaited.
Collapse
Affiliation(s)
- Rupesh Raina
- Department of Nephrology, Cleveland Clinic Akron General/Akron Nephrology Associates, Akron, Ohio, USA.,Akron Children's Hospital, Akron, Ohio, USA
| | | | - Ronith Chakraborty
- Department of Nephrology, Cleveland Clinic Akron General/Akron Nephrology Associates, Akron, Ohio, USA
| | - Nikhil Nair
- Case Western Reserve University, Cleveland, Ohio, USA
| | - Haikoo Shah
- Northeast Ohio Medical University, Rootstown, Ohio, USA
| | - Vinod Krishnappa
- Department of Nephrology, Cleveland Clinic Akron General/Akron Nephrology Associates, Akron, Ohio, USA.,Northeast Ohio Medical University, Rootstown, Ohio, USA
| | | |
Collapse
|
149
|
Brill AL, Ehrlich BE. Polycystin 2: A calcium channel, channel partner, and regulator of calcium homeostasis in ADPKD. Cell Signal 2019; 66:109490. [PMID: 31805375 DOI: 10.1016/j.cellsig.2019.109490] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 11/30/2019] [Accepted: 12/01/2019] [Indexed: 01/26/2023]
Abstract
Polycystin 2 (PC2) is one of two main protein types responsible for the underlying etiology of autosomal dominant polycystic kidney disease (ADPKD), the most prevalent monogenic renal disease in the world. This debilitating and currently incurable condition is caused by loss-of-function mutations in PKD2 and PKD1, the genes encoding for PC2 and Polycystin 1 (PC1), respectively. Two-hit mutation events in these genes lead to renal cyst formation and eventual kidney failure, the main hallmarks of ADPKD. Though much is known concerning the physiological consequences and dysfunctional signaling mechanisms resulting from ADPKD development, to best understand the requirement of PC2 in maintaining organ homeostasis, it is important to recognize how PC2 acts under normal conditions. As such, an array of work has been performed characterizing the endogenous function of PC2, revealing it to be a member of the transient receptor potential (TRP) channel family of proteins. As a TRP protein, PC2 is a nonselective, cation-permeant, calcium-sensitive channel expressed in all tissue types, where it localizes primarily on the endoplasmic reticulum (ER), primary cilia, and plasma membrane. In addition to its channel function, PC2 interacts with and acts as a regulator of a number of other channels, ultimately further affecting intracellular signaling and leading to dysfunction in its absence. In this review, we describe the biophysical and physiological properties of PC2 as a cation channel and modulator of intracellular calcium channels, along with how these properties are altered in ADPKD.
Collapse
Affiliation(s)
- Allison L Brill
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Barbara E Ehrlich
- Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA; Department of Pharmacology, Yale University, New Haven, CT, USA.
| |
Collapse
|
150
|
Geng X, Zhong D, Su L, Lin Z, Yang B. Preventive and therapeutic effect of Ganoderma lucidum on kidney injuries and diseases. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2019; 87:257-276. [PMID: 32089235 DOI: 10.1016/bs.apha.2019.10.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ganoderma lucidum (G. lucidum, Lingzhi) is a well-known Chinese traditional medicine to improve health and to treat numerous diseases for over 2000 years in Asian countries. G. lucidum has the abundant chemical components such as triterpenes and polysaccharides, which have various biological activities including anti-oxidation, anti-inflammation, anti-liver disorders, anti-tumor growth and metastasis, etc. Recently, many lines of studies have elucidated the therapeutic effects of G. lucidum and its extractions on various acute kidney injury (AKI) and chronic kidney disease (CKD) pathogenesis, including autosomal dominant polycystic kidney disease, diabetic nephropathy, renal proximal tubular cell oxidative damage and fibrotic process, renal ischemia reperfusion injury, cisplatin-induced renal injury, adriamycin-induced nephropathy, chronic proteinuric renal diseases, etc. Clinical researches also showed potent anti-renal disease bioactivities of G. lucidum. In this chapter, we review experimental and clinical researches and provide comprehensive insights into the renoprotective effects of G. lucidum. In recent years, renal diseases have gradually aroused attention on account of their booming prevalence worldwide and lack of effective therapies. Although the complicated pathogenesis of kidney diseases, such as acute kidney injury (AKI) and chronic kidney diseases (CKD) have been intensively studied. The morbidity and mortality of AKI and CKD still rise continuously. Thanks to the conventional experience and the multi-target characteristics, natural products have been increasingly recognized as an alternative source for treating renal diseases.
Collapse
Affiliation(s)
- Xiaoqiang Geng
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Dandan Zhong
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Limin Su
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Zhibin Lin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China
| | - Baoxue Yang
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Pharmacology, School of Basic Medical Sciences, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing, China.
| |
Collapse
|