1
|
Zagorec N, Calamel A, Delaporte M, Olinger E, Orr S, Sayer JA, Pillay VG, Denommé-Pichon AS, Mau-Them FT, Nambot S, Faivre L, Ars E, Torra R, Ong AC, Devuyst O, Perico N, Després AM, Lemoine H, de Fallois J, Brousse R, Hummel A, Knebelmann B, Maisonneuve N, Halbritter J, Le Meur Y, Audrézet MP, Cornec-Le Gall E. Clinical Spectrum and Prognosis of Atypical Autosomal Dominant Polycystic Kidney Disease Caused by Monoallelic Pathogenic Variants of IFT140. Am J Kidney Dis 2024:S0272-6386(24)01126-0. [PMID: 39732359 DOI: 10.1053/j.ajkd.2024.10.009] [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: 04/08/2024] [Revised: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 12/30/2024]
Abstract
RATIONALE & OBJECTIVE Monoallelic predicted Loss-of-Function (pLoF) variants in IFT140 have recently been associated with an autosomal dominant polycystic kidney disease (ADPKD)-like phenotype. This study sought to enhance the characterization of this phenotype. STUDY DESIGN Case series. SETTING & PARTICIPANTS Seventy-five among 2797 European individuals with ADPKD-like phenotypes who underwent genetic testing that revealed pLoF IFT140-variants. FINDINGS The 75 individuals (median age 56 years, 53.3% females) were from 61 families and were found to have 41 different monoallelic pLoF IFT140-variants. The majority of individuals presented with large, exophytic kidney cysts (median [range] total kidney volume 688 ml [201-4139]), and 90.2% were classified using the Mayo Imaging Classification as Mayo Class 2A. Arterial hypertension was present in 50.7% of the individuals (median [range] age at diagnosis 59 years [29-73]). Only one patient developed kidney failure (at age 69 years). A significant difference in age-adjusted eGFR between male and female patients was observed (P<0.001). 56.3% of the individuals over the age of 60 years had an eGFR less than 60ml/min/1.73m2. The estimated genetic prevalence of monoallelic pLoF IFT140 variants was 19.76 (95%CI=18.8-20.7) and 27.89 (95%CI=23.8-31.9) per 10,000 in the Genome Aggregation Database and the 100,000 Genomes Project (100kG), respectively. CyKD (ICD-10 Q61) was associated with pLoF IFT140 variants (P=2.9x10-9, OR=5.6 (3.3-9.2)) only in 100kG. STUDY LIMITATIONS Retrospective study; younger patients and patients with milder forms of IFT140-related CyKD may not be diagnosed. CONCLUSIONS Individuals with monoallelic IFT140 pLoF variants are likely to develop kidney cysts atypical of classical ADPKD and generally have a favorable kidney prognosis.
Collapse
Affiliation(s)
- Nikola Zagorec
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de référence MARHEA, CHRU Brest, Brest, France
| | - Alizée Calamel
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de référence MARHEA, CHRU Brest, Brest, France
| | - Margaux Delaporte
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de référence MARHEA, CHRU Brest, Brest, France
| | - Eric Olinger
- Center for Human Genetics, Cliniques Universitaires Saint-Luc, UCLouvain, Brussels, Belgium
| | - Sarah Orr
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, United Kingdom
| | - John A Sayer
- Biosciences Institute, Newcastle University, Central Parkway, Newcastle Upon Tyne, United Kingdom; The Newcastle upon Tyne Hospitals NHS Foundation Trust, Renal Services, Freeman Road, Newcastle Upon Tyne, United Kingdom; NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Vignesh-Guru Pillay
- Unité Fonctionnelle Innovation en Diagnostic Génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Anne Sophie Denommé-Pichon
- Unité Fonctionnelle Innovation en Diagnostic Génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Frederic Tran Mau-Them
- Unité Fonctionnelle Innovation en Diagnostic Génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France
| | - Sophie Nambot
- INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté, Dijon, France
| | - Laurence Faivre
- INSERM UMR1231 GAD, Genetics of Developmental Disorders, Université de Bourgogne-Franche-Comté, Dijon, France; Centre de Génétique et Centre de Référence Anomalies du Développement et Syndromes Malformatifs de l'Interrégion Est, Centre Hospitalier Universitaire Dijon, Dijon, France; Fédération Hospitalo-Universitaire Médecine Translationnelle et Anomalies du Développement (TRANSLAD), CHU Dijon, Dijon, France
| | - Elisabet Ars
- Molecular Biology Laboratory, Fundació Puigvert, Instituto de Recerca Sant Pau (R Sant Pau), RICORS2040, Barcelona, Spain
| | - Roser Torra
- Department of Nephrology, Fundació Puigvert, Instituto de Recerca Sant Pau (R Sant Pau), Universitat Autónoma de Barcelona, Medicine Department, RICORS2040, Barcelona, Spain
| | - Albert Cm Ong
- Kidney Genetics Group, Academic Nephrology, Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield, UK; Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Olivier Devuyst
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Noberto Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Aurore Michel Després
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de référence MARHEA, CHRU Brest, Brest, France
| | - Hugo Lemoine
- Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | | | - Romain Brousse
- University of Brest, Inserm, UMR 1078, Génétique, Génomique fonctionelle et Biotechnologies, Brest, France
| | - Aurélie Hummel
- Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Bertrand Knebelmann
- Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Nathalie Maisonneuve
- Sorbonne Université, INSERM UMRS 1155, Nephrology Department, Assistance Publique Hopitaux De Paris, Hopital Tenon, Paris, France
| | - Jan Halbritter
- Department of Nephrology, Necker Hospital, Assistance Publique-Hopitaux de Paris, Paris, France
| | - Yannick Le Meur
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de référence MARHEA, CHRU Brest, Brest, France; Service de Néphrologie-Dialyse, Centre Hospitalier de Valenciennes, Valenciennes, France
| | - Marie-Pierre Audrézet
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy; Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium
| | - Emilie Cornec-Le Gall
- Service de Néphrologie, Hémodialyse et Transplantation Rénale, Centre de référence MARHEA, CHRU Brest, Brest, France; Institut de Recherche Expérimentale et Clinique (IREC), UCLouvain, Brussels, Belgium.
| |
Collapse
|
2
|
Kleene SJ. Hyperosmolality activates polycystin-2 and TRPM4 in renal primary cilium. Pflugers Arch 2024:10.1007/s00424-024-03050-8. [PMID: 39688695 DOI: 10.1007/s00424-024-03050-8] [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: 05/29/2024] [Revised: 10/09/2024] [Accepted: 12/01/2024] [Indexed: 12/18/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a debilitating disease characterized by renal cysts. It arises from mutations in proteins expressed in part in the primary cilia of renal epithelial cells. One of these, polycystin-2 (PC2), is an ion-conducting channel. To date, ion channels in the cilium have only been characterized in standard normosmolar external solutions, but the osmolality of the renal filtrate bathing the cilia varies widely. Here I report that urine, which better represents the filtrate, activates a large cation-conducting current in the cilia. With defined external solutions, hyperosmolality through addition of urea, NaCl, or D-mannitol activates a similar current. Most but not all of this current is conducted through TRPM4 channels. It is greatly reduced by internal MgATP or 9-phenanthrol, which inhibit TRPM4, or by shRNA knockdown of TRPM4. However, part of the current activated by urea conducts Ca2+ through channels that remain to be identified. External hyperosmolality also greatly increases the activity of ciliary PC2 channels; this is the first physiological stimulus identified for these channels. Possibilities are discussed for the mechanisms of channel activation and the roles for these activities in regulatory volume increase and cystogenesis.
Collapse
Affiliation(s)
- Steven J Kleene
- Department of Pharmacology, Physiology, and Neurobiology, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
3
|
Ha K, Loeb GB, Park M, Gupta M, Akiyama Y, Argiris J, Pinedo A, Park CH, Brandes N, Ritu F, Ye CJ, Reiter JF, Delling M. ADPKD-Causing Missense Variants in Polycystin-1 Disrupt Cell Surface Localization or Polycystin Channel Function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.04.570035. [PMID: 38106161 PMCID: PMC10723288 DOI: 10.1101/2023.12.04.570035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the leading monogenic cause of kidney failure and affects millions of people worldwide. Despite the prevalence of this monogenic disorder, our limited mechanistic understanding of ADPKD has hindered therapeutic development. Here, we successfully developed bioassays that functionally classify missense variants in polycystin-1 (PC1). Strikingly, ADPKD pathogenic missense variants cluster into two major categories: 1) those that disrupt polycystin cell surface localization or 2) those that attenuate polycystin ion channel activity. We found that polycystin channels with defective surface localization could be rescued with a small molecule. We propose that small-molecule-based strategies to improve polycystin cell surface localization and channel function will be effective therapies for ADPKD patients.
Collapse
|
4
|
Solano AS, Lavanderos B, Metwally E, Earley S. Transient Receptor Potential Channels in Vascular Mechanotransduction. Am J Hypertens 2024:hpae134. [PMID: 39579078 DOI: 10.1093/ajh/hpae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 09/17/2024] [Accepted: 10/04/2024] [Indexed: 11/25/2024] Open
Abstract
Transmural pressure and shear stress are mechanical forces that profoundly affect the smooth muscle cells (SMCs) comprising the vascular wall and the endothelial cells (ECs) lining the lumen. Pressure and flow are detected by mechanosensors in these cells and translated into appropriate responses to regulate blood pressure and flow. This review focuses on the role of the transient receptor potential (TRP) superfamily of cation channels in this process. We discuss how specific members of the TRP superfamily (TRPC6, TRPM4, TRPV1, TRPV4, and TRPP1) regulate the resting membrane and intracellular Ca2+ levels in SMCs and ECs to promote changes in vascular tone in response to intraluminal pressure and shear stress. Although TRP channels participate in vascular mechanotransduction, little evidence supports their intrinsic mechanosensitivity. Therefore, we also examine the evidence exploring the force-sensitive signal transduction pathways acting upstream of vascular TRP channels. Understanding the interplay between mechanosensors, force-induced signaling cascades, and TRP channels holds promise for the development of targeted therapies for diseases caused by vascular dysfunction.
Collapse
Affiliation(s)
- Alfredo Sanchez Solano
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Boris Lavanderos
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| | - Elsayed Metwally
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Scott Earley
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
5
|
Busch T, Neubauer B, Schmitt L, Cascante I, Knoblich L, Wegehaupt O, Schöler F, Tholen S, Hofherr A, Schell C, Schilling O, Westermann L, Köttgen M. The role of the co-chaperone DNAJB11 in polycystic kidney disease: Molecular mechanisms and cellular origin of cyst formation. FASEB J 2024; 38:e70162. [PMID: 39530576 DOI: 10.1096/fj.202401763r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 10/18/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in PKD1 and PKD2, encoding polycystin-1 (PC1) and polycystin-2 (PC2), which are required for the regulation of the renal tubular diameter. Loss of polycystin function results in cyst formation. Atypical forms of ADPKD are caused by mutations in genes encoding endoplasmic reticulum (ER)-resident proteins through mechanisms that are not well understood. Here, we investigate the function of DNAJB11, an ER co-chaperone associated with atypical ADPKD. We generated mouse models with constitutive and conditional Dnajb11 inactivation and Dnajb11-deficient renal epithelial cells to investigate the mechanism underlying autosomal dominant inheritance, the specific cell types driving cyst formation, and molecular mechanisms underlying DNAJB11-dependent polycystic kidney disease. We show that biallelic loss of Dnajb11 causes cystic kidney disease and fibrosis, mirroring human disease characteristics. In contrast to classical ADPKD, cysts predominantly originate from proximal tubules. Cyst formation begins in utero and the timing of Dnajb11 inactivation strongly influences disease severity. Furthermore, we identify impaired PC1 cleavage as a potential mechanism underlying DNAJB11-dependent cyst formation. Proteomic analysis of Dnajb11- and Pkd1-deficient cells reveals common and distinct pathways and dysregulated proteins, providing a foundation to better understand phenotypic differences between different forms of ADPKD.
Collapse
Affiliation(s)
- Tilman Busch
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Björn Neubauer
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Lars Schmitt
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Isabel Cascante
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Luise Knoblich
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Oliver Wegehaupt
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felix Schöler
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Stefan Tholen
- Department of Pathology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Alexis Hofherr
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Christoph Schell
- Department of Pathology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Oliver Schilling
- Department of Pathology, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Lukas Westermann
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
| | - Michael Köttgen
- Department of Medicine IV - Nephrology and Primary Care, Faculty of Medicine and Medical Center, University of Freiburg, Freiburg, Germany
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| |
Collapse
|
6
|
Wang S, Kang Y, Xie H. PKD2: An Important Membrane Protein in Organ Development. Cells 2024; 13:1722. [PMID: 39451240 PMCID: PMC11506562 DOI: 10.3390/cells13201722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 10/11/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
PKD2 was first identified as the pathogenic protein for autosomal dominant polycystic kidney disease (ADPKD) and is widely recognized as an ion channel. Subsequent studies have shown that PKD2 is widely expressed in various animal tissues and plays a crucial role in tissue and organ development. Additionally, PKD2 is conserved from single-celled organisms to vertebrates. Here, we provide an overview of recent advances in the function of PKD2 in key model animals, focusing on the establishment of left-right organ asymmetry, renal homeostasis, cardiovascular development, and signal transduction in reproduction and mating. We specifically focus on the roles of PKD2 in development and highlight future prospects for PKD2 research.
Collapse
Affiliation(s)
- Shuo Wang
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Yunsi Kang
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Haibo Xie
- MoE Key Laboratory of Evolution and Marine Biodiversity, Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao 266003, China; (S.W.); (Y.K.)
- College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| |
Collapse
|
7
|
Nakatani S, Kawano H, Sato M, Hoshino J, Nishio S, Miura K, Sekine A, Suwabe T, Hidaka S, Kataoka H, Ishikawa E, Shimazu K, Uchiyama K, Fujimaru T, Moriyama T, Kurashige M, Shimabukuro W, Hattanda F, Kimura T, Ushio Y, Manabe S, Watanabe H, Mitobe M, Seta K, Shimada Y, Kai H, Katayama K, Ichikawa D, Hayashi H, Hanaoka K, Mochizuki T, Nakanishi K, Tsuchiya K, Horie S, Isaka Y, Muto S. Protocol for the nationwide registry of patients with polycystic kidney disease: japanese national registry of PKD (JRP). Clin Exp Nephrol 2024; 28:1004-1015. [PMID: 38734869 DOI: 10.1007/s10157-024-02509-3] [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: 02/19/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
BACKGROUND Autosomal dominant polycystic kidney disease (ADPKD) and autosomal recessive polycystic kidney disease (ARPKD) are major genetic polycystic kidney diseases that can progress to end-stage kidney disease (ESKD). Longitudinal data on the clinical characteristics associated with clinical outcomes in polycystic kidney disease (PKD), including the development of ESKD and cardiovascular disease (CVD) are lacking in Japan. To address this unmet need the authors are establishing a novel, web-based, Nationwide Cohort Registry Study-the Japanese Registry of PKD (JRP). METHODS The JRP is a prospective cohort study for ADPKD (aim to recruit n = 1000 patients), and both a retrospective and prospective study for ARPKD (aim to recruit n = 100). In the prospective registry, patients will be followed-up for 10 years every 6 months and 12 months for patients with ADPKD and ARPKD, respectively. Data collection will be recorded on Research Electronic Data Capture (REDCap) starting on April 1, 2024, with recruitment ending on March 31, 2029. (jRCT 1030230618). RESULTS Data to be collected include: baseline data, demographics, diagnostic and genetic information, radiological and laboratory findings, and therapeutic interventions. During follow-up, clinical events such as development of ESKD, hospitalization, occurrence of extra kidney complications including CVD events, and death will be recorded, as well as patient-reported health-related quality of life for patients with ADPKD. CONCLUSIONS The JRP is the first nationwide registry study for patients with ADPKD and ARPKD in Japan, providing researchers with opportunities to advance knowledge and treatments for ADPKD and ARPKD, and to inform disease management and future clinical practice.
Collapse
Affiliation(s)
- Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Haruna Kawano
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Advanced Informatics for Genetic Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Mai Sato
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Saori Nishio
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | | | | | - Sumi Hidaka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Hiroshi Kataoka
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Eiji Ishikawa
- Department of Nephrology, Saiseikai Matsusaka General Hospital, Mie, Japan
| | - Keiji Shimazu
- Department of Nephrology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Kiyotaka Uchiyama
- Department of Nephrology, International University of Health and Welfare Narita Hospital, Chiba, Japan
| | - Takuya Fujimaru
- Department of Nephrology, St. Luke's International Hospital, Tokyo, Japan
| | - Tomofumi Moriyama
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Mahiro Kurashige
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Wataru Shimabukuro
- Department of Child Health and Welfare (Pediatrics), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Fumihiko Hattanda
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Tomoki Kimura
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yusuke Ushio
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Shun Manabe
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hirofumi Watanabe
- Division of Clinical Nephrology and Rheumatology, Kidney Research Center, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Michihiro Mitobe
- Department of Nephrology, Takeda General Hospital, Fukushima, Japan
| | - Koichi Seta
- Department of Nephrology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Yosuke Shimada
- Intelligent Systems Laboratory, SECOM CO., LTD, Mitaka, Tokyo, Japan
- Infection Control Science, Juntendo University Graduate School, Bunkyo, Tokyo, Japan
| | - Hirayasu Kai
- Ibaraki Clinical Education and Training Center, Institute of Medicine, University of Tsukuba Ibaraki, Tsukuba, Japan
| | - Kan Katayama
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Mie, Japan
| | - Daisuke Ichikawa
- Department of Nephrology and Hypertension, St Marianna University School of Medicine, Kanagawa, Japan
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine, Aichi, Japan
| | - Kazushige Hanaoka
- Department of General Internal Medicine, School of Medicine, Daisan Hospital The Jikei University, Tokyo, Japan
| | | | - Koichi Nakanishi
- Department of Child Health and Welfare (Pediatrics), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Ken Tsuchiya
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Shigeo Horie
- Department of Urology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Satoru Muto
- Department of Urology, Juntendo University Nerima Hospital, 3-1-10, Takanodai, Nerima-ku, Tokyo, 177-8521, Japan.
| |
Collapse
|
8
|
Zhang F, Mehta H, Choudhary HH, Islam R, Hanafy KA. TRPV4 Channel in Neurological Disease: from Molecular Mechanisms to Therapeutic Potential. Mol Neurobiol 2024:10.1007/s12035-024-04518-5. [PMID: 39333347 DOI: 10.1007/s12035-024-04518-5] [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: 05/30/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024]
Abstract
Transient Receptor Potential Vanilloid 4 (TRPV4) is a non-selective cation channel with pivotal roles in various physiological processes, including osmosensitivity, mechanosensation, neuronal development, vascular tone regulation, and bone homeostasis in human bodies. Recent studies have made significant progress in understanding the structure and functional role of TRPV4, shedding light on its involvement in pathological processes, particularly in the realm of neurological diseases. Here, we aim to provide a comprehensive exploration of the multifaceted contributions of TRPV4 to neurological diseases, spanning its intricate molecular mechanisms to its potential as a target for therapeutic interventions. We delve into the structural and functional attributes of TRPV4, scrutinize its expression profile, and elucidate the possible mechanisms through which it participates in the pathogenesis of neurological disorders. Furthermore, we discussed recent years' progress in therapeutic strategies aimed at harnessing TRPV4 for the treatment of these diseases. These insights will provide a basis for understanding and designing modality-specific pharmacological agents to treat TRPV4-associated disorders.
Collapse
Affiliation(s)
- Feng Zhang
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hritik Mehta
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Rezwanul Islam
- Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper University Health Care, Camden, NJ, USA
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Khalid A Hanafy
- Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation at Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper Neurological Institute Center for Neuroinflammation, Cooper Medical School at Rowan University, Camden, NJ, USA.
| |
Collapse
|
9
|
Nitta K, Kataoka H, Manabe S, Makabe S, Akihisa T, Ushio Y, Seki M, Tsuchiya K, Hoshino J, Mochizuki T. Association of hyperphosphatemia with renal prognosis in patients with autosomal dominant polycystic kidney disease. Clin Exp Nephrol 2024:10.1007/s10157-024-02568-6. [PMID: 39322826 DOI: 10.1007/s10157-024-02568-6] [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: 05/18/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
BACKGROUND Serum phosphate (P) levels are generally lower in autosomal dominant polycystic kidney disease (ADPKD) than in other kidney disorders, potentially masking the clinical significance of hyperphosphatemia. This study aimed to determine if serum P levels can predict renal outcomes in ADPKD patients. METHODS We included 235 patients with ADPKD who were not taking drugs to treat hyperphosphatemia. Survival analysis was performed for the renal outcome of a 50% reduction in estimated glomerular filtration rate or initiation of renal replacement therapy. RESULTS Multivariable Cox regression analyses revealed that serum P (1 mg/dL increase, HR = 2.03, P < 0.0001) was a significant risk factor for kidney disease progression. Similarly, hyperphosphatemia (P > 3.5 mg/dL, HR = 2.05; P > 4.0 mg/dL, HR = 1.90; P > 4.5 mg/dL, HR = 2.78; P > 5.0 mg/dL, HR = 27.22) was significantly associated with renal prognosis. Kaplan-Meier analysis showed significantly lower kidney survival rates in patients with P > 3.5 mg/dL than in those without hyperphosphatemia (log-rank test, P < 0.0001), and similar Kaplan-Meier analysis results were found for P > 4.0 mg/dL, P > 4.5 mg/dL, and P > 5.0 mg/dL. The 2 year kidney survival rate for ADPKD patients with P > 3.5 mg/dL was 66.7% overall and 41.4% in those with stage 4-5 CKD. For patients with P > 4.0 mg/dL, the survival rate dropped to 46.8% overall and 28.2% in those with stage 4-5 CKD, indicating a very poor prognosis. CONCLUSION Hyperphosphatemia was associated with renal prognosis in patients with ADPKD. In these patients, attention should be paid to even mild serum P elevation of > 3.5 or > 4.0 mg/dL.
Collapse
Affiliation(s)
- Kosaku Nitta
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Hiroshi Kataoka
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan.
| | - Shun Manabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Shiho Makabe
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Taro Akihisa
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Yusuke Ushio
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Momoko Seki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Ken Tsuchiya
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
| | - Toshio Mochizuki
- Department of Nephrology, Tokyo Women's Medical University, 8-1 Kawada-Cho, Shinjuku-Ku, Tokyo, 162-8666, Japan
- PKD Nephrology Clinic, Tokyo, Japan
| |
Collapse
|
10
|
Palomero OE, DeCaen PG. ADPKD variants in the PKD2 pore helix cause structural collapse of the gate and distinct forms of channel dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612744. [PMID: 39314384 PMCID: PMC11419077 DOI: 10.1101/2024.09.12.612744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
PKD2 is a member of the polycystin subfamily of transient receptor potential (TRP) ion channel subunits which traffic and function in primary cilia organelle membranes. Millions of individuals carry pathogenic genetic variants in PKD2 that cause a life-threatening condition called autosomal dominant polycystic kidney disease (ADPKD). Although ADPKD is a common monogenetic disorder, there is no drug cure or available therapeutics which address the underlying channel dysregulation. Furthermore, the structural and mechanistic impact of most disease-causing variants are uncharacterized. Using direct cilia electrophysiology, cryogenic electron microscopy (cryo-EM), and super resolution imaging, we have discovered mechanistic differences in channel dysregulation caused by three germline missense variants located in PKD2's pore helix 1. Variant C632R reduces protein thermal stability, resulting in impaired channel assembly and abolishes primary cilia trafficking. In contrast, variants F629S and R638C retain native cilia trafficking, but exhibit gating defects. Resolved cryo-EM structures (2.7-3.2Å) of the variants indicate loss of critical pore helix interactions and precipitate allosteric collapse of the channels inner gate. Results demonstrate how ADPKD-causing these mutations have divergent and ranging impacts on PKD2 function, despite their shared structural proximity. These unexpected findings underscore the need for mechanistic characterization of polycystin variants, which may guide rational drug development of ADPKD therapeutics. Regarding polycystin nomenclature The revised and current IUPHAR/BPS nomenclature creates ambiguity regarding the genetic identity of the polycystin family members of transient receptor potential ion channels (TRPP), especially when cross-referencing manuscripts that describe subunits using the former system 1 . Traditionally, the products of polycystin genes (e.g., PKD2) are referred to as polycystin proteins (e.g., polycystin-2). For simplicity and to prevent confusion, we will refer to the polycystin gene name rather than differentiating gene and protein with separate names- a nomenclature we have recently outlined (Annual Reviews in Physiology, Esarte Palomero et al. 2023) 2.
Collapse
|
11
|
De Paolis E, Raspaglio G, Ciferri N, Zangrilli I, Ricciardi Tenore C, Urbani A, Ferraro PM, Minucci A, Concolino P. Single-Base Substitution Causing Dual-Exon Skipping Event in PKD2 Gene: Unusual Molecular Finding from Exome Sequencing in a Patient with Autosomal Dominant Polycystic Kidney Disease. J Clin Med 2024; 13:4682. [PMID: 39200828 PMCID: PMC11355194 DOI: 10.3390/jcm13164682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Background: Pathogenic variants in the Polycystic Kidney Disease 2 (PKD2) gene are associated with Autosomal Dominant Polycystic Kidney Disease (ADPKD) in approximately 30% of cases. In recent years, the high-throughput sequencing techniques have significantly increased the number of variants identified in affected patients. Here, we described the peculiar effect of a PKD2 splicing variant, the c.1717-2A>G, identified in an Italian male patient with ADPKD. This variant led to the unusual and rare skipping of two consecutive exons, causing a large in-frame deletion. Methods: The genetic evaluation of the patient was performed using the Next-Generation Sequencing (NGS) assay Clinical Exome Solution® (SOPHiA Genetics). Bioinformatics analysis was performed using the SOPHiA DDM platform (SOPHiA Genetics). Prediction of pathogenicity was carried out by integrating several in silico tools. RNA evaluation was performed to test the effect of the variant on the PKD2 splicing using a Reverse-Transcription PCR coupled with cDNA sequencing. Results: NGS revealed the presence of the PKD2 c.1717-2A>G variant that lies in the canonical splice site of intron 7. This rare variant was predicted to have a significant impact on the splicing, proved by the RNA-based analysis. We identified the presence of a transcript characterised by the simultaneous skipping of exons 8 and 9, with a retained reading frame and the merging of exons 7-10. Conclusions: We described for the first time a dual-exon skip event related to the presence of a single-base substitution in the PKD2 gene in an ADPKD-affected patient. We assumed that the molecular basis of such a rare mechanism lies in the specific order of intron removal. The finding represents novel evidence of an alternative and unusual splicing mechanism in the PKD2 gene, adding insights to the pathogenesis of the ADPKD.
Collapse
Affiliation(s)
- Elisa De Paolis
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (G.R.); (I.Z.); (C.R.T.); (A.M.)
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Giuseppina Raspaglio
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (G.R.); (I.Z.); (C.R.T.); (A.M.)
- Division of Oncological Gynecology, Department of Women’s and Children’s Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Nunzia Ciferri
- Nephrology Unit, Departement of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| | - Ilaria Zangrilli
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (G.R.); (I.Z.); (C.R.T.); (A.M.)
| | - Claudio Ricciardi Tenore
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (G.R.); (I.Z.); (C.R.T.); (A.M.)
| | - Andrea Urbani
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Pietro Manuel Ferraro
- Section of Nephrology, Department of Medicine, Università degli Studi di Verona, 37127 Verona, Italy;
| | - Angelo Minucci
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (G.R.); (I.Z.); (C.R.T.); (A.M.)
| | - Paola Concolino
- Departmental Unit of Molecular and Genomic Diagnostics, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy; (E.D.P.); (G.R.); (I.Z.); (C.R.T.); (A.M.)
- Clinical Chemistry, Biochemistry and Molecular Biology Operations (UOC), Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy;
| |
Collapse
|
12
|
Koyano T, Onishi K, Matsuyama M, Fukushima M, Kume K. Extracellular calcium promotes internalization and degradation of the fission yeast TRP-like calcium ion channel Pkd2. MICROPUBLICATION BIOLOGY 2024; 2024:10.17912/micropub.biology.001265. [PMID: 39165565 PMCID: PMC11333956 DOI: 10.17912/micropub.biology.001265] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/03/2024] [Accepted: 07/31/2024] [Indexed: 08/22/2024]
Abstract
The correct localization of proteins is linked to their cellular function. The Schizosaccharomyces pombe Pkd2 localizes to the endoplasmic reticulum and plasma membrane. Here we investigate the behavior of Pkd2 in response to calcium. Pkd2-GFP, normally enriched at the cell ends, is reduced from the plasma membrane by CaCl 2 addition, while cytoplasmic dots and free GFP are increased. This suggests that Pkd2 is internalized and degraded in response to extracellular CaCl 2 . This internalization is partially suppressed by treatment with an Arp2/3 inhibitor, CK-666. Our data provide new insights into the relationship between Pkd2 internalization and calcium response.
Collapse
Affiliation(s)
- Takayuki Koyano
- Division of Cell Biology, Shigei Medical Research Institute, Okayama, Okayama, Japan
| | - Kaori Onishi
- Division of Cell Biology, Shigei Medical Research Institute, Okayama, Okayama, Japan
| | - Makoto Matsuyama
- Division of Molecular Genetics, Shigei Medical Research Institute, Okayama, Okayama, Japan
| | | | - Kazunori Kume
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
- Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| |
Collapse
|
13
|
Staudner T, Geiges L, Khamseekaew J, Sure F, Korbmacher C, Ilyaskin AV. Disease-associated missense mutations in the pore loop of polycystin-2 alter its ion channel function in a heterologous expression system. J Biol Chem 2024; 300:107574. [PMID: 39009345 PMCID: PMC11630642 DOI: 10.1016/j.jbc.2024.107574] [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: 02/16/2024] [Revised: 06/20/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024] Open
Abstract
Polycystin-2 (PC2) is mutated in ∼15% of patients with autosomal dominant polycystic kidney disease (ADPKD). PC2 belongs to the family of transient receptor potential (TRP) channels and can function as a homotetramer. We investigated whether three disease-associated mutations (F629S, C632R, or R638C) localized in the channel's pore loop alter ion channel properties of human PC2 expressed in Xenopus laevis oocytes. Expression of wild-type (WT) PC2 typically resulted in small but measurable Na+ inward currents in the absence of extracellular divalent cations. These currents were no longer observed when individual pore mutations were introduced in WT PC2. Similarly, Na+ inward currents mediated by the F604P gain-of-function (GOF) PC2 construct (PC2 F604P) were abolished by each of the three pore mutations. In contrast, when the mutations were introduced in another GOF construct, PC2 L677A N681A, only C632R had a complete loss-of-function effect, whereas significant residual Na+ inward currents were observed with F629S (∼15%) and R638C (∼30%). Importantly, the R638C mutation also abolished the Ca2+ permeability of PC2 L677A N681A and altered its monovalent cation selectivity. To elucidate the molecular mechanisms by which the R638C mutation affects channel function, molecular dynamics (MD) simulations were used in combination with functional experiments and site-directed mutagenesis. Our findings suggest that R638C stabilizes ionic interactions between Na+ ions and the selectivity filter residue D643. This probably explains the reduced monovalent cation conductance of the mutant channel. In summary, our data support the concept that altered ion channel properties of PC2 contribute to the pathogenesis of ADPKD.
Collapse
Affiliation(s)
- Tobias Staudner
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Cellular and Molecular Physiology, Erlangen, Germany
| | - Linda Geiges
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Cellular and Molecular Physiology, Erlangen, Germany
| | - Juthamas Khamseekaew
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Cellular and Molecular Physiology, Erlangen, Germany
| | - Florian Sure
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Cellular and Molecular Physiology, Erlangen, Germany
| | - Christoph Korbmacher
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Cellular and Molecular Physiology, Erlangen, Germany
| | - Alexandr V Ilyaskin
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Institute of Cellular and Molecular Physiology, Erlangen, Germany.
| |
Collapse
|
14
|
Rezi CK, Aslanyan MG, Diwan GD, Cheng T, Chamlali M, Junger K, Anvarian Z, Lorentzen E, Pauly KB, Afshar-Bahadori Y, Fernandes EF, Qian F, Tosi S, Christensen ST, Pedersen SF, Strømgaard K, Russell RB, Miner JH, Mahjoub MR, Boldt K, Roepman R, Pedersen LB. DLG1 functions upstream of SDCCAG3 and IFT20 to control ciliary targeting of polycystin-2. EMBO Rep 2024; 25:3040-3063. [PMID: 38849673 PMCID: PMC11239879 DOI: 10.1038/s44319-024-00170-1] [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: 12/11/2023] [Revised: 05/08/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024] Open
Abstract
Polarized vesicular trafficking directs specific receptors and ion channels to cilia, but the underlying mechanisms are poorly understood. Here we describe a role for DLG1, a core component of the Scribble polarity complex, in regulating ciliary protein trafficking in kidney epithelial cells. Conditional knockout of Dlg1 in mouse kidney causes ciliary elongation and cystogenesis, and cell-based proximity labeling proteomics and fluorescence microscopy show alterations in the ciliary proteome upon loss of DLG1. Specifically, the retromer-associated protein SDCCAG3, IFT20, and polycystin-2 (PC2) are reduced in the cilia of DLG1-deficient cells compared to control cells. This phenotype is recapitulated in vivo and rescuable by re-expression of wild-type DLG1, but not a Congenital Anomalies of the Kidney and Urinary Tract (CAKUT)-associated DLG1 variant, p.T489R. Finally, biochemical approaches and Alpha Fold modelling suggest that SDCCAG3 and IFT20 form a complex that associates, at least indirectly, with DLG1. Our work identifies a key role for DLG1 in regulating ciliary protein composition and suggests that ciliary dysfunction of the p.T489R DLG1 variant may contribute to CAKUT.
Collapse
Affiliation(s)
- Csenge K Rezi
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Mariam G Aslanyan
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gaurav D Diwan
- BioQuant, Heidelberg University, Heidelberg, Germany
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Tao Cheng
- Department of Medicine (Nephrology Division) and Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| | - Mohamed Chamlali
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Katrin Junger
- Institute for Ophthalmic Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Zeinab Anvarian
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Esben Lorentzen
- Department of Molecular Biology and Genetics - Protein Science, Aarhus University, Aarhus, Denmark
| | - Kleo B Pauly
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | | | - Eduardo Fa Fernandes
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Feng Qian
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sébastien Tosi
- Danish BioImaging Infrastructure Image Analysis Core Facility (DBI-INFRA IACF), University of Copenhagen, Copenhagen, Denmark
| | | | - Stine F Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Robert B Russell
- BioQuant, Heidelberg University, Heidelberg, Germany
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Jeffrey H Miner
- Department of Medicine (Nephrology Division) and Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| | - Moe R Mahjoub
- Department of Medicine (Nephrology Division) and Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| | - Karsten Boldt
- Institute for Ophthalmic Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Ronald Roepman
- Department of Human Genetics, Research Institute for Medical Innovation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Lotte B Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
15
|
Chowdhury P, Sinha D, Poddar A, Chetluru M, Chen Q. The Mechanosensitive Pkd2 Channel Modulates the Recruitment of Myosin II and Actin to the Cytokinetic Contractile Ring. J Fungi (Basel) 2024; 10:455. [PMID: 39057340 PMCID: PMC11277609 DOI: 10.3390/jof10070455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Cytokinesis, the last step in cell division, separates daughter cells through mechanical force. This is often through the force produced by an actomyosin contractile ring. In fission yeast cells, the ring helps recruit a mechanosensitive ion channel, Pkd2, to the cleavage furrow, whose activation by membrane tension promotes calcium influx and daughter cell separation. However, it is unclear how the activities of Pkd2 may affect the actomyosin ring. Here, through both microscopic and genetic analyses of a hypomorphic pkd2 mutant, we examined the potential role of this essential gene in assembling the contractile ring. The pkd2-81KD mutation significantly increased the counts of the type II myosin heavy chain Myo2 (+18%), its regulatory light chain Rlc1 (+37%) and actin (+100%) molecules in the ring, compared to the wild type. Consistent with a regulatory role of Pkd2 in the ring assembly, we identified a strong negative genetic interaction between pkd2-81KD and the temperature-sensitive mutant myo2-E1. The pkd2-81KD myo2-E1 cells often failed to assemble a complete contractile ring. We conclude that Pkd2 modulates the recruitment of type II myosin and actin to the contractile ring, suggesting a novel calcium-dependent mechanism regulating the actin cytoskeletal structures during cytokinesis.
Collapse
Affiliation(s)
| | | | | | | | - Qian Chen
- Department of Biological Sciences, The University of Toledo, 2801 Bancroft St, Toledo, OH 43606, USA; (P.C.); (D.S.); (M.C.)
| |
Collapse
|
16
|
Laboyrie SL, Svensson MK, Josemans S, Sigvant B, Rotmans JI, Welander G. Vascular Access Outcomes in Patients with Autosomal Dominant Polycystic Kidney Disease. KIDNEY360 2024; 5:877-885. [PMID: 38985981 PMCID: PMC11219118 DOI: 10.34067/kid.0000000000000453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/19/2024] [Indexed: 05/03/2024]
Abstract
Key Points More patients with autosomal dominant polycystic kidney disease received their first intervention to re-establish vascular access patency. Patients with autosomal dominant polycystic kidney disease do not require differential monitoring and treatment of hemodialysis vascular access. Background Autosomal dominant polycystic kidney disease (ADPKD) is a leading hereditary cause of ESKD, often using hemodialysis as a form of RRT. Patients with ADPKD may also present with extrarenal manifestations, including arterial aneurysms. The gold standard for hemodialysis access is an arteriovenous vascular access (VA), such as arteriovenous fistulas (AVFs) or arteriovenous grafts (AVGs). However, limitations, such as low VA flow and inadequate AVF outward remodeling, affect VA utilization. This study aimed to explore whether ADPKD affects patency rates of AVFs/AVGs in comparison with other underlying ESKD causes. Methods We conducted a retrospective cohort study using data from the Swedish Renal Registry from 2011 to 2020, with follow-up until 2022. We included 496 patients with ADPKD and 4321 propensity score–matched controls. VA patency rates of patients with ADPKD were compared with those of non-ADPKD patients using Kaplan–Meier survival curves and Mantel–Cox log-rank test. Interventions to maintain or restore patency were also analyzed. Results Patients with ADPKD constituted 8.0% of all patients, with a higher proportion in the pre-ESKD phase during VA creation (51.6% versus 40.6%). No significant differences were observed in primary, postcannulation primary, secondary, or functional patency between patients with ADPKD and non-ADPKD patients. However, more VAs were ligated in patients with ADPKD (10.5% versus 7.7%, P = 0.03), and they underwent more first interventions to re-establish flow (49.4% versus 41.9%, P = 0.02). Conclusions These findings suggest that AVF/AVG patency remains comparable in patients with ESKD with or without ADPKD, and VA monitoring and treatment strategies for patients with ADPKD should align with those for individuals with other ESKD causes.
Collapse
Affiliation(s)
- Suzanne L. Laboyrie
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Maria K. Svensson
- Department of Medical Sciences Renal Medicine, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Centre, Uppsala University, Uppsala, Sweden
| | - Sabine Josemans
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Birgitta Sigvant
- Department of Surgical Sciences, Center of Clinical Research, Uppsala University, Uppsala, Sweden
| | - Joris I. Rotmans
- Department of Internal Medicine, Leiden University Medical Centre, Leiden, The Netherlands
| | - Gunilla Welander
- Department of Medical Sciences Renal Medicine, Uppsala University, Uppsala, Sweden
- Center of Clinical Research, Region Värmland, Sweden
| |
Collapse
|
17
|
Di Mise A, Caplan MJ, Valenti G. Editorial: Molecular mechanisms underlying polycystic kidney disease: from the smallest bricks to the big scenario. Front Mol Biosci 2024; 11:1429206. [PMID: 38836108 PMCID: PMC11149700 DOI: 10.3389/fmolb.2024.1429206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 06/06/2024] Open
Affiliation(s)
- Annarita Di Mise
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| | - Michael J Caplan
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
18
|
Schönauer R, Sierks D, Boerrigter M, Jawaid T, Caroff L, Audrezet MP, Friedrich A, Shaw M, Degenhardt J, Forberger M, de Fallois J, Bläker H, Bergmann C, Gödiker J, Schindler P, Schlevogt B, Müller RU, Berg T, Patterson I, Griffiths WJ, Sayer JA, Popp B, Torres VE, Hogan MC, Somlo S, Watnick TJ, Nevens F, Besse W, Cornec-Le Gall E, Harris PC, Drenth JPH, Halbritter J. Sex, Genotype, and Liver Volume Progression as Risk of Hospitalization Determinants in Autosomal Dominant Polycystic Liver Disease. Gastroenterology 2024; 166:902-914. [PMID: 38101549 DOI: 10.1053/j.gastro.2023.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/04/2023] [Accepted: 12/10/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND & AIMS Autosomal dominant polycystic liver disease is a rare condition with a female preponderance, based mainly on pathogenic variants in 2 genes, PRKCSH and SEC63. Clinically, autosomal dominant polycystic liver disease is characterized by vast heterogeneity, ranging from asymptomatic to highly symptomatic hepatomegaly. To date, little is known about the prediction of disease progression at early stages, hindering clinical management, genetic counseling, and the design of randomized controlled trials. To improve disease prognostication, we built a consortium of European and US centers to recruit the largest cohort of patients with PRKCSH and SEC63 liver disease. METHODS We analyzed an international multicenter cohort of 265 patients with autosomal dominant polycystic liver disease harboring pathogenic variants in PRKCSH or SEC63 for genotype-phenotype correlations, including normalized age-adjusted total liver volumes and polycystic liver disease-related hospitalization (liver event) as primary clinical end points. RESULTS Classifying individual total liver volumes into predefined progression groups yielded predictive risk discrimination for future liver events independent of sex and underlying genetic defects. In addition, disease severity, defined by age at first liver event, was considerably more pronounced in female patients and patients with PRKCSH variants than in those with SEC63 variants. A newly developed sex-gene score was effective in distinguishing mild, moderate, and severe disease, in addition to imaging-based prognostication. CONCLUSIONS Both imaging and clinical genetic scoring have the potential to inform patients about the risk of developing symptomatic disease throughout their lives. The combination of female sex, germline PRKCSH alteration, and rapid total liver volume progression is associated with the greatest odds of polycystic liver disease-related hospitalization.
Collapse
Affiliation(s)
- Ria Schönauer
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin (corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin), Berlin, Germany; Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Dana Sierks
- Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany; Department of Pediatric Surgery, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Melissa Boerrigter
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tabinda Jawaid
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Lea Caroff
- University of Brest, Institut National de la Santé et de la Recherche Médicale, UMR 1078, Génétique, Génomique Fonctionnelle et Biotechnologies, Brest, France; Centre Hospitalier Universitaire Brest, Service de Néphrologie, Centre de Référence Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Brest, France
| | - Marie-Pierre Audrezet
- Centre Hospitalier Universitaire Brest, Service de Génétique Moléculaire, Brest, France
| | - Anja Friedrich
- Medizinische Genetik Mainz, Limbach Genetics, Mainz, Germany
| | - Melissa Shaw
- Departments of Internal Medicine and Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Jan Degenhardt
- Department 2 of Internal Medicine, University of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany
| | - Mirjam Forberger
- Department of Pathology, University of Leipzig Medical Center, Leipzig, Germany
| | - Jonathan de Fallois
- Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany
| | - Hendrik Bläker
- Department of Pathology, University of Leipzig Medical Center, Leipzig, Germany
| | | | - Juliana Gödiker
- Department of Internal Medicine B, University Hospital Münster, Münster, Germany
| | | | - Bernhard Schlevogt
- Department of Internal Medicine B, University Hospital Münster, Münster, Germany; Department of Gastroenterology, Medical Center Osnabrück, Osnabrück, Germany
| | - Roman-U Müller
- Department 2 of Internal Medicine, University of Cologne, Faculty of Medicine, University Hospital Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Thomas Berg
- Division of Hepatology, Department of Medicine II, University of Leipzig Medical Center, Leipzig, Germany
| | - Ilse Patterson
- Department of Radiology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - William J Griffiths
- Department of Hepatology, Cambridge Liver Unit, Cambridge University Hospitals, Cambridge, United Kingdom
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Renal Services, Newcastle upon Tyne National Health Service Foundation Trust, Newcastle upon Tyne, United Kingdom; National Institute for Health Research Newcastle Biomedical Research Centre, Newcastle upon Tyne, United Kingdom
| | - Bernt Popp
- Berlin Institute of Health at Charité, Universitätsmedizin Berlin, Center of Functional Genomics, Berlin, Germany
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Marie C Hogan
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Stefan Somlo
- Departments of Internal Medicine and Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Terry J Watnick
- Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland
| | - Frederik Nevens
- Department of Hepatology and Liver Transplantation Unit, University Hospitals Katholieke Universiteit Leuven, Leuven, Belgium
| | - Whitney Besse
- Departments of Internal Medicine and Nephrology, Yale University School of Medicine, New Haven, Connecticut
| | - Emilie Cornec-Le Gall
- University of Brest, Institut National de la Santé et de la Recherche Médicale, UMR 1078, Génétique, Génomique Fonctionnelle et Biotechnologies, Brest, France; Centre Hospitalier Universitaire Brest, Service de Néphrologie, Centre de Référence Maladies Rénales Héréditaires de l'Enfant et de l'Adulte, Brest, France
| | - Peter C Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Joost P H Drenth
- Department of Gastroenterology and Hepatology, Radboud University Medical Center, Nijmegen, The Netherlands.
| | - Jan Halbritter
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin (corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin), Berlin, Germany; Division of Nephrology, Department of Internal Medicine, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
19
|
Yen PW, Chen YA, Wang W, Mao FS, Chao CT, Chiang CK, Lin SH, Tarng DC, Chiu YW, Wu MJ, Chen YC, Kao JTW, Wu MS, Lin CL, Huang JW, Hung KY. The screening, diagnosis, and management of patients with autosomal dominant polycystic kidney disease: A national consensus statement from Taiwan. Nephrology (Carlton) 2024; 29:245-258. [PMID: 38462235 DOI: 10.1111/nep.14287] [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: 10/10/2023] [Revised: 01/29/2024] [Accepted: 02/25/2024] [Indexed: 03/12/2024]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited cause of end-stage kidney disease (ESKD) worldwide. Guidelines for the diagnosis and management of ADPKD in Taiwan remains unavailable. In this consensus statement, we summarize updated information on clinical features of international and domestic patients with ADPKD, followed by suggestions for optimal diagnosis and care in Taiwan. Specifically, counselling for at-risk minors and reproductive issues can be important, including ethical dilemmas surrounding prenatal diagnosis and pre-implantation genetic diagnosis. Studies reveal that ADPKD typically remains asymptomatic until the fourth decade of life, with symptoms resulting from cystic expansion with visceral compression, or rupture. The diagnosis can be made based on a detailed family history, followed by imaging studies (ultrasound, computed tomography, or magnetic resonance imaging). Genetic testing is reserved for atypical cases mostly. Common tools for prognosis prediction include total kidney volume, Mayo classification and PROPKD/genetic score. Screening and management of complications such as hypertension, proteinuria, urological infections, intracranial aneurysms, are also crucial for improving outcome. We suggest that the optimal management strategies of patients with ADPKD include general medical care, dietary recommendations and ADPKD-specific treatments. Key points include rigorous blood pressure control, dietary sodium restriction and Tolvaptan use, whereas the evidence for somatostatin analogues and mammalian target of rapamycin (mTOR) inhibitors remains limited. In summary, we outline an individualized care plan emphasizing careful monitoring of disease progression and highlight the need for shared decision-making among these patients.
Collapse
Affiliation(s)
- Pao-Wen Yen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Yung-An Chen
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Wei Wang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Fang-Sheng Mao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Chia-Ter Chao
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Min-Sheng General Hospital, Taoyuan City, Taiwan
| | - Chih-Kang Chiang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Shih-Hua Lin
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Der-Cherng Tarng
- Division of Nephrology, Department of Internal Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Wen Chiu
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Ju Wu
- Division of Nephrology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung City, Taiwan
| | - Yung-Chang Chen
- Kidney Research Center, Department of Nephrology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Juliana Tze-Wah Kao
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
- Division of Nephrology, Department of Internal Medicine, Fu-Jen Catholic University Hospital, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Mai-Szu Wu
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chun-Liang Lin
- Division of Nephrology, Department of Internal Medicine, Chia-Yi Chang Gung Memorial Hospital, Chia-Yi County, Taiwan
| | - Jenq-Wen Huang
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
| | - Kuan-Yu Hung
- Division of Nephrology, Department of Internal Medicine, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan
- Division of Nephrology, Department of Internal Medicine, Taipei Medical University-Shuang-Ho Hospital, Ministry of Health and Welfare, New Taipei City, Taiwan
| |
Collapse
|
20
|
Mbiakop UC, Jaggar JH. Vascular polycystin proteins in health and disease. Microcirculation 2024; 31:e12834. [PMID: 37823335 PMCID: PMC11009377 DOI: 10.1111/micc.12834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/13/2023]
Abstract
PKD1 (polycystin 1) and PKD2 (polycystin 2) are expressed in a variety of different cell types, including arterial smooth muscle and endothelial cells. PKD1 is a transmembrane domain protein with a large extracellular N-terminus that is proposed to act as a mechanosensor and receptor. PKD2 is a member of the transient receptor potential (TRP) channel superfamily which is also termed TRPP1. Mutations in the genes which encode PKD1 and PKD2 lead to autosomal dominant polycystic kidney disease (ADPKD). ADPKD is one of the most prevalent monogenic disorders in humans and is associated with extrarenal and vascular complications, including hypertension. Recent studies have uncovered mechanisms of activation and physiological functions of PKD1 and PKD2 in arterial smooth muscle and endothelial cells. It has also been found that PKD function is altered in the vasculature during ADPKD and hypertension. We will summarize this work and discuss future possibilities for this area of research.
Collapse
Affiliation(s)
- Ulrich C. Mbiakop
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38163
| | - Jonathan H. Jaggar
- Department of Physiology, University of Tennessee Health Science Center, Memphis TN 38163
| |
Collapse
|
21
|
Chowdhury P, Sinha D, Poddar A, Chetluru M, Chen Q. The mechanosensitive Pkd2 channel modulates the recruitment of myosin II and actin to the cytokinetic contractile ring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.15.575753. [PMID: 38293176 PMCID: PMC10827123 DOI: 10.1101/2024.01.15.575753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Cytokinesis, the last step in cell division, separate daughter cells through the force produced by an actomyosin contractile ring assembled at the equatorial plane. In fission yeast cells, the ring helps recruit a mechanosensitive ion channel Pkd2 to the cleavage furrow, whose activation by membrane tension promotes calcium influx and daughter cell separation. However, it is unclear how the activities of Pkd2 may affect the actomyosin ring. Here, through both microscopic and genetic analyses of a hypomorphic mutant of the essential pkd2 gene, we examine its potential role in assembling and constricting the contractile ring. The pkd2-81KD mutation significantly increased the number of type II myosin heavy chain Myo2 (+20%), its regulatory light chain Rlc1 (+37%) and actin (+20%) molecules in the ring, compared to the wild type. Consistent with a regulatory role of Pkd2 in the ring assembly, we identified a strong negative genetic interaction between pkd2-81KD and the temperature-sensitive mutant myo2-E1 . The pkd2-81KD myo2-E1 cells often failed to assemble a complete contractile ring. We conclude that Pkd2 modulates the recruitment of type II myosin and actin to the contractile ring, suggesting a novel calcium- dependent mechanism regulating the actin cytoskeletal structures during cytokinesis.
Collapse
|
22
|
Vishy CE, Thomas C, Vincent T, Crawford DK, Goddeeris MM, Freedman BS. Genetics of cystogenesis in base-edited human organoids reveal therapeutic strategies for polycystic kidney disease. Cell Stem Cell 2024; 31:537-553.e5. [PMID: 38579684 PMCID: PMC11325856 DOI: 10.1016/j.stem.2024.03.005] [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: 02/21/2023] [Revised: 12/19/2023] [Accepted: 03/08/2024] [Indexed: 04/07/2024]
Abstract
In polycystic kidney disease (PKD), microscopic tubules expand into macroscopic cysts. Among the world's most common genetic disorders, PKD is inherited via heterozygous loss-of-function mutations but is theorized to require additional loss of function. To test this, we establish human pluripotent stem cells in allelic series representing four common nonsense mutations, using CRISPR base editing. When differentiated into kidney organoids, homozygous mutants spontaneously form cysts, whereas heterozygous mutants (original or base corrected) express no phenotype. Using these, we identify eukaryotic ribosomal selective glycosides (ERSGs) as PKD therapeutics enabling ribosomal readthrough of these same nonsense mutations. Two different ERSGs not only prevent cyst initiation but also limit growth of pre-formed cysts by partially restoring polycystin expression. Furthermore, glycosides accumulate in cyst epithelia in organoids and mice. Our findings define the human polycystin threshold as a surmountable drug target for pharmacological or gene therapy interventions, with relevance for understanding disease mechanisms and future clinical trials.
Collapse
Affiliation(s)
- Courtney E Vishy
- Division of Nephrology, Department of Medicine, Institute for Stem Cell and Regenerative Medicine, and Kidney Research Institute, University of Washington, Seattle, WA 98109, USA
| | - Chardai Thomas
- Division of Nephrology, Department of Medicine, Institute for Stem Cell and Regenerative Medicine, and Kidney Research Institute, University of Washington, Seattle, WA 98109, USA
| | - Thomas Vincent
- Division of Nephrology, Department of Medicine, Institute for Stem Cell and Regenerative Medicine, and Kidney Research Institute, University of Washington, Seattle, WA 98109, USA
| | - Daniel K Crawford
- Eloxx Pharmaceuticals, Inc., 950 Winter Street, Waltham, MA 02451, USA
| | | | - Benjamin S Freedman
- Division of Nephrology, Department of Medicine, Institute for Stem Cell and Regenerative Medicine, and Kidney Research Institute, University of Washington, Seattle, WA 98109, USA; Plurexa, 1209 6th Ave. N., Seattle, WA 98109, USA.
| |
Collapse
|
23
|
Lee WC, Cheng BC, Lee CT, Liao SC. Update on the Application of Ultrasonography in Understanding Autosomal Dominant Polycystic Kidney Disease. J Med Ultrasound 2024; 32:110-115. [PMID: 38882609 PMCID: PMC11175384 DOI: 10.4103/jmu.jmu_77_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 06/18/2024] Open
Abstract
With an estimated prevalence of 1 in 1000 individuals globally, autosomal dominant polycystic kidney disease (ADPKD) stands as the most prevalent inherited renal disorder. Ultrasonography (US) is the most widely used imaging modality in the diagnosis and monitoring of ADPKD. This review discusses the role of US in the evaluation of ADPKD, including its diagnostic accuracy, limitations, and recent advances. An overview of the pathophysiology and clinical manifestations of ADPKD has also been provided. Furthermore, the potential of US as a noninvasive tool for the assessment of disease progression and treatment response is examined. Overall, US remains an essential tool for the management of ADPKD, and ongoing research efforts are aimed at improving its diagnostic and prognostic capabilities.
Collapse
Affiliation(s)
- Wen-Chin Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ben-Chung Cheng
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chien-Te Lee
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Municipal Feng-Shan Hospital, Kaohsiung, Taiwan
| | - Shang-Chih Liao
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Division of Nephrology, Department of Internal Medicine, Kaohsiung Municipal Feng-Shan Hospital, Kaohsiung, Taiwan
| |
Collapse
|
24
|
Zhang J, Wang Y, Zhao Y, Liu F. A new atypical splice mutation in PKD2 leading to autosomal dominant polycystic kidney disease in a Chinese family. Singapore Med J 2024; 65:229-234. [PMID: 34749493 PMCID: PMC11132625 DOI: 10.11622/smedj.2021162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 07/25/2021] [Indexed: 02/05/2023]
Abstract
INTRODUCTION Autosomal dominant polycystic kidney disease (ADPKD) is a very common hereditary renal disorder. Mutations in PKD1 and PKD2 , identified as disease-causing genes, account for 85% and 15% of the ADPKD cases, respectively. METHODS In this study, the mutation analysis of polycystic kidney disease (PKD) genes was performed in a Chinese family with suspected ADPKD using targeted clinical exome sequencing (CES). The candidate pathogenic variants were further tested by using Sanger sequencing and validated for co-segregation. In addition, reverse transcription-polymerase chain reaction (RT-PCR) was performed to test for abnormal splicing and assess its potential pathogenicity. RESULTS A novel atypical splicing mutation that belongs to unclassified variants (UCVs), IVS6+5G>C, was identified in three family members by CES and was shown to co-segregate only with the affected individuals. The RT-PCR revealed the abnormal splicing of exon 6, which thus caused truncating mutation. These findings suggested that the atypical splice site alteration, IVS6+5G>C, in the PKD2 gene was the potential pathogenic mutation leading to ADPKD in this Chinese family. CONCLUSION The data available in this study provided strong evidence that IVS6+5G>C is the potential pathogenic mutation for ADPKD. In addition, our findings emphasised the significance of functional analysis of UCVs and genotype-phenotype correlation in ADPKD.
Collapse
Affiliation(s)
- Junlin Zhang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yiting Wang
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| | - Yingwang Zhao
- Joy Orient Translational Medicine Research Center Co Ltd, Beijing, China
| | - Fang Liu
- Division of Nephrology, West China Hospital of Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
25
|
Huang J, Tao H, Chen J, Shen Y, Lei J, Pan J, Yan C, Yan N. Structure-guided discovery of protein and glycan components in native mastigonemes. Cell 2024; 187:1733-1744.e12. [PMID: 38552612 DOI: 10.1016/j.cell.2024.02.037] [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: 09/23/2023] [Revised: 01/07/2024] [Accepted: 02/27/2024] [Indexed: 04/02/2024]
Abstract
Mastigonemes, the hair-like lateral appendages lining cilia or flagella, participate in mechanosensation and cellular motion, but their constituents and structure have remained unclear. Here, we report the cryo-EM structure of native mastigonemes isolated from Chlamydomonas at 3.0 Å resolution. The long stem assembles as a super spiral, with each helical turn comprising four pairs of anti-parallel mastigoneme-like protein 1 (Mst1). A large array of arabinoglycans, which represents a common class of glycosylation in plants and algae, is resolved surrounding the type II poly-hydroxyproline (Hyp) helix in Mst1. The EM map unveils a mastigoneme axial protein (Mstax) that is rich in heavily glycosylated Hyp and contains a PKD2-like transmembrane domain (TMD). Mstax, with nearly 8,000 residues spanning from the intracellular region to the distal end of the mastigoneme, provides the framework for Mst1 assembly. Our study provides insights into the complexity of protein and glycan interactions in native bio-architectures.
Collapse
Affiliation(s)
- Junhao Huang
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Hui Tao
- MOE Key Laboratory of Protein Sciences, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jikun Chen
- MOE Key Laboratory of Protein Sciences, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yang Shen
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jianlin Lei
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Junmin Pan
- MOE Key Laboratory of Protein Sciences, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, Shandong, China.
| | - Chuangye Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Nieng Yan
- Beijing Frontier Research Center for Biological Structures, State Key Laboratory of Membrane Biology, Tsinghua-Peking Joint Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute of Bio-Architecture and Bio-Interactions (IBABI), Shenzhen Medical Academy of Research and Translation (SMART), Shenzhen, Guangdong 518107, China.
| |
Collapse
|
26
|
Rezi CK, Aslanyan MG, Diwan GD, Cheng T, Chamlali M, Junger K, Anvarian Z, Lorentzen E, Pauly KB, Afshar-Bahadori Y, Fernandes EFA, Qian F, Tosi S, Christensen ST, Pedersen SF, Strømgaard K, Russell RB, Miner JH, Mahjoub MR, Boldt K, Roepman R, Pedersen LB. DLG1 functions upstream of SDCCAG3 and IFT20 to control ciliary targeting of polycystin-2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.10.566524. [PMID: 37987012 PMCID: PMC10659422 DOI: 10.1101/2023.11.10.566524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Polarized vesicular trafficking directs specific receptors and ion channels to cilia, but the underlying mechanisms are poorly understood. Here we describe a role for DLG1, a core component of the Scribble polarity complex, in regulating ciliary protein trafficking in kidney epithelial cells. Conditional knockout of Dlg1 in mouse kidney caused ciliary elongation and cystogenesis, and cell-based proximity labelling proteomics and fluorescence microscopy showed alterations in the ciliary proteome upon loss of DLG1. Specifically, the retromer-associated protein SDCCAG3, IFT20 and polycystin-2 (PC2) were reduced in cilia of DLG1 deficient cells compared to control cells. This phenotype was recapitulated in vivo and rescuable by re-expression of wildtype DLG1, but not a Congenital Anomalies of the Kidney and Urinary Tract (CAKUT)-associated DLG1 variant, p.T489R. Finally, biochemical approaches and Alpha Fold modelling suggested that SDCCAG3 and IFT20 form a complex that associates, at least indirectly, with DLG1. Our work identifies a key role for DLG1 in regulating ciliary protein composition and suggests that ciliary dysfunction of the p.T489R DLG1 variant may contribute to CAKUT.
Collapse
Affiliation(s)
- Csenge K. Rezi
- Department of Biology, University of Copenhagen, Denmark
| | - Mariam G. Aslanyan
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Gaurav D. Diwan
- BioQuant, Heidelberg University, Heidelberg, Germany
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Tao Cheng
- Department of Medicine (Nephrology Division) and Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| | | | - Katrin Junger
- Institute for Ophthalmic Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | | | - Esben Lorentzen
- Department of Molecular Biology and Genetics - Protein Science, Aarhus University, Denmark
| | - Kleo B. Pauly
- Department of Biology, University of Copenhagen, Denmark
| | | | - Eduardo F. A. Fernandes
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Feng Qian
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Sébastien Tosi
- Danish BioImaging Infrastructure Image Analysis Core Facility (DBI-INFRA IACF), University of Copenhagen, Denmark
| | | | | | - Kristian Strømgaard
- Center for Biopharmaceuticals, Department of Drug Design and Pharmacology, University of Copenhagen, Denmark
| | - Robert B. Russell
- BioQuant, Heidelberg University, Heidelberg, Germany
- Biochemistry Center (BZH), Heidelberg University, Heidelberg, Germany
| | - Jeffrey H. Miner
- Department of Medicine (Nephrology Division) and Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| | - Moe R. Mahjoub
- Department of Medicine (Nephrology Division) and Department of Cell Biology and Physiology, Washington University, St Louis, MO, USA
| | - Karsten Boldt
- Institute for Ophthalmic Research, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Ronald Roepman
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | | |
Collapse
|
27
|
Righini M, Mancini R, Busutti M, Buscaroli A. Autosomal Dominant Polycystic Kidney Disease: Extrarenal Involvement. Int J Mol Sci 2024; 25:2554. [PMID: 38473800 DOI: 10.3390/ijms25052554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disorder, but kidneys are not the only organs involved in this systemic disorder. Individuals with the condition may display additional manifestations beyond the renal system, involving the liver, pancreas, and brain in the context of cystic manifestations, while involving the vascular system, gastrointestinal tract, bones, and cardiac valves in the context of non-cystic manifestations. Despite kidney involvement remaining the main feature of the disease, thanks to longer survival, early diagnosis, and better management of kidney-related problems, a new wave of complications must be faced by clinicians who treated patients with ADPKD. Involvement of the liver represents the most prevalent extrarenal manifestation and has growing importance in the symptom burden and quality of life. Vascular abnormalities are a key factor for patients' life expectancy and there is still debate whether to screen or not to screen all patients. Arterial hypertension is often the earliest onset symptom among ADPKD patients, leading to frequent cardiovascular complications. Although cardiac valvular abnormalities are a frequent complication, they rarely lead to relevant problems in the clinical history of polycystic patients. One of the newest relevant aspects concerns bone disorders that can exert a considerable influence on the clinical course of these patients. This review aims to provide the "state of the art" among the extrarenal manifestation of ADPKD.
Collapse
Affiliation(s)
- Matteo Righini
- Nephrology and Dialysis Unit, Santa Maria delle Croci Hospital, AUSL Romagna, 48121 Ravenna, Italy
- Nephrology, Dialysis and Transplantation Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Raul Mancini
- Nephrology, Dialysis and Transplantation Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Marco Busutti
- Nephrology, Dialysis and Transplantation Unit, IRCCS Azienda Ospedaliero Universitaria di Bologna, 40138 Bologna, Italy
| | - Andrea Buscaroli
- Nephrology and Dialysis Unit, Santa Maria delle Croci Hospital, AUSL Romagna, 48121 Ravenna, Italy
| |
Collapse
|
28
|
Flockerzi V, Fakler B. TR(i)P Goes On: Auxiliary TRP Channel Subunits? Circ Res 2024; 134:346-350. [PMID: 38359093 DOI: 10.1161/circresaha.123.323178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/28/2023] [Indexed: 02/17/2024]
Abstract
Transient receptor potential (TRP) cation channels are a diverse family of channels whose members play prominent roles as cellular sensors and effectors. The important role of TRP channels (and mechanosensitive piezo channels) in the complex interaction of our senses with the environment was underlined by the award of the Nobel Prize in Physiology or Medicine to 2 pioneers in this field, David Julius and Ardem Patapoutian. There are many competent and comprehensive reviews on many aspects of the TRP channels, and there is no intention to expand on them. Rather, after an introduction to the nomenclature, the molecular architecture of native TRP channel/protein complexes in vivo will be summarized using TRP channels of the canonical transient receptor potential subfamily as an example. This molecular architecture provides the basis for the signatures of native canonical transient receptor potential currents and their control by endogenous modulators and potential drugs.
Collapse
Affiliation(s)
- Veit Flockerzi
- Experimental and Clinical Pharmacology and Toxicology, Saarland University, Homburg, Germany (V.F.)
| | - Bernd Fakler
- Institute of Physiology, Faculty of Medicine, University of Freiburg, Freiburg, Germany (B.F.)
| |
Collapse
|
29
|
Thompson WS, Babayev SN, McGowan ML, Kattah AG, Wick MJ, Bendel-Stenzel EM, Chebib FT, Harris PC, Dahl NK, Torres VE, Hanna C. State of the Science and Ethical Considerations for Preimplantation Genetic Testing for Monogenic Cystic Kidney Diseases and Ciliopathies. J Am Soc Nephrol 2024; 35:235-248. [PMID: 37882743 PMCID: PMC10843344 DOI: 10.1681/asn.0000000000000253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 10/03/2023] [Indexed: 10/27/2023] Open
Abstract
There is a broad phenotypic spectrum of monogenic polycystic kidney diseases (PKDs). These disorders often involve cilia-related genes and lead to the development of fluid-filled cysts and eventual kidney function decline and failure. Preimplantation genetic testing for monogenic (PGT-M) disorders has moved into the clinical realm. It allows prospective parents to avoid passing on heritable diseases to their children, including monogenic PKD. The PGT-M process involves embryo generation through in vitro fertilization, with subsequent testing of embryos and selective transfer of those that do not harbor the specific disease-causing variant(s). There is a growing body of literature supporting the success of PGT-M for autosomal-dominant and autosomal-recessive PKD, although with important technical limitations in some cases. This technology can be applied to many other types of monogenic PKD and ciliopathies despite the lack of existing reports in the literature. PGT-M for monogenic PKD, like other forms of assisted reproductive technology, raises important ethical questions. When considering PGT-M for kidney diseases, as well as the potential to avoid disease in future generations, there are regulatory and ethical considerations. These include limited government regulation and unstandardized consent processes, potential technical errors, high cost and equity concerns, risks associated with pregnancy for mothers with kidney disease, and the impact on all involved in the process, including the children who were made possible with this technology.
Collapse
Affiliation(s)
- Whitney S. Thompson
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Biomedical Ethics Research Program, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Division of Neonatal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Samir N. Babayev
- Division of Reproductive Endocrinology and Infertility, Mayo Clinic, Rochester, Minnesota
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | - Michelle L. McGowan
- Center for Individualized Medicine, Mayo Clinic, Rochester, Minnesota
- Biomedical Ethics Research Program, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, Minnesota
| | - Andrea G. Kattah
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Myra J. Wick
- Department of Clinical Genomics, Mayo Clinic, Rochester, Minnesota
- Department of Obstetrics and Gynecology, Mayo Clinic, Rochester, Minnesota
| | | | - Fouad T. Chebib
- Division of Nephrology and Hypertension, Mayo Clinic, Jacksonville, Florida
| | - Peter C. Harris
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Neera K. Dahl
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Vicente E. Torres
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Christian Hanna
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
- Division of Pediatric Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
30
|
Yasinoglu SA, Kuipers TB, Suidgeest E, van der Weerd L, Mei H, Baelde HJ, Peters DJM. Transcriptomic profiling of Polycystic Kidney Disease identifies paracrine factors in the early cyst microenvironment. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166987. [PMID: 38070582 DOI: 10.1016/j.bbadis.2023.166987] [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: 08/23/2023] [Revised: 11/30/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023]
Abstract
Initial cysts that are formed upon Pkd1 loss in mice impose persistent stress on surrounding tissue and trigger a cystic snowball effect, in which local aberrant PKD-related signaling increases the likelihood of new cyst formation, ultimately leading to accelerated disease progression. Although many pathways have been associated with PKD progression, the knowledge of early changes near initial cysts is limited. To perform an unbiased analysis of transcriptomic alterations in the cyst microenvironment, microdomains were collected from kidney sections of iKsp-Pkd1del mice with scattered Pkd1-deletion using Laser Capture Microdissection. These microdomains were defined as F4/80-low cystic, representing early alterations in the cyst microenvironment, F4/80-high cystic, with more advanced alterations, or non-cystic. RNA sequencing and differential gene expression analysis revealed 953 and 8088 dysregulated genes in the F4/80-low and F4/80-high cyst microenvironment, respectively, when compared to non-cystic microdomains. In the early cyst microenvironment, several injury-repair, growth, and tissue remodeling-related pathways were activated, accompanied by mild metabolic changes. In the more advanced F4/80-high microdomains, these pathways were potentiated and the metabolism was highly dysregulated. Upstream regulator analysis revealed a series of paracrine factors with increased activity in the early cyst microenvironment, including TNFSF12 and OSM. In line with the upstream regulator analysis, TWEAK and Oncostatin-M promoted cell proliferation and inflammatory gene expression in renal epithelial cells and fibroblasts in vitro. Collectively, our data provide an overview of molecular alterations that specifically occur in the cyst microenvironment and identify paracrine factors that may mediate early and advanced alterations in the cyst microenvironment.
Collapse
Affiliation(s)
- Sevtap A Yasinoglu
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas B Kuipers
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Ernst Suidgeest
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Louise van der Weerd
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans J Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Dorien J M Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
31
|
Kalot R, Sentell Z, Kitzler TM, Torban E. Primary cilia and actin regulatory pathways in renal ciliopathies. FRONTIERS IN NEPHROLOGY 2024; 3:1331847. [PMID: 38292052 PMCID: PMC10824913 DOI: 10.3389/fneph.2023.1331847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/20/2023] [Indexed: 02/01/2024]
Abstract
Ciliopathies are a group of rare genetic disorders caused by defects to the structure or function of the primary cilium. They often affect multiple organs, leading to brain malformations, congenital heart defects, and anomalies of the retina or skeletal system. Kidney abnormalities are among the most frequent ciliopathic phenotypes manifesting as smaller, dysplastic, and cystic kidneys that are often accompanied by renal fibrosis. Many renal ciliopathies cause chronic kidney disease and often progress to end-stage renal disease, necessitating replacing therapies. There are more than 35 known ciliopathies; each is a rare hereditary condition, yet collectively they account for a significant proportion of chronic kidney disease worldwide. The primary cilium is a tiny microtubule-based organelle at the apex of almost all vertebrate cells. It serves as a "cellular antenna" surveying environment outside the cell and transducing this information inside the cell to trigger multiple signaling responses crucial for tissue morphogenesis and homeostasis. Hundreds of proteins and unique cellular mechanisms are involved in cilia formation. Recent evidence suggests that actin remodeling and regulation at the base of the primary cilium strongly impacts ciliogenesis. In this review, we provide an overview of the structure and function of the primary cilium, focusing on the role of actin cytoskeleton and its regulators in ciliogenesis. We then describe the key clinical, genetic, and molecular aspects of renal ciliopathies. We highlight what is known about actin regulation in the pathogenesis of these diseases with the aim to consider these recent molecular findings as potential therapeutic targets for renal ciliopathies.
Collapse
Affiliation(s)
- Rita Kalot
- Department of Medicine and Department of Physiology, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Center, Montreal, QC, Canada
| | - Zachary Sentell
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Thomas M. Kitzler
- Department of Human Genetics, McGill University, Montreal, QC, Canada
- McGill University Health Center, Montreal, QC, Canada
| | - Elena Torban
- Department of Medicine and Department of Physiology, McGill University, Montreal, QC, Canada
- The Research Institute of the McGill University Health Center, Montreal, QC, Canada
| |
Collapse
|
32
|
Kita T, Chiba K, Wang J, Nakagawa A, Niwa S. Comparative analysis of two Caenorhabditis elegans kinesins KLP-6 and UNC-104 reveals a common and distinct activation mechanism in kinesin-3. eLife 2024; 12:RP89040. [PMID: 38206323 PMCID: PMC10945585 DOI: 10.7554/elife.89040] [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] [Indexed: 01/12/2024] Open
Abstract
Kinesin-3 is a family of microtubule-dependent motor proteins that transport various cargos within the cell. However, the mechanism underlying kinesin-3 activations remains largely elusive. In this study, we compared the biochemical properties of two Caenorhabditis elegans kinesin-3 family proteins, KLP-6 and UNC-104. Both KLP-6 and UNC-104 are predominantly monomeric in solution. As previously shown for UNC-104, non-processive KLP-6 monomer is converted to a processive motor when artificially dimerized. We present evidence that releasing the autoinhibition is sufficient to trigger dimerization of monomeric UNC-104 at nanomolar concentrations, which results in processive movement of UNC-104 on microtubules, although it has long been thought that enrichment in the phospholipid microdomain on cargo vesicles is required for the dimerization and processive movement of UNC-104. In contrast, KLP-6 remains to be a non-processive monomer even when its autoinhibition is unlocked, suggesting a requirement of other factors for full activation. By examining the differences between KLP-6 and UNC-104, we identified a coiled-coil domain called coiled-coil 2 (CC2) that is required for the efficient dimerization and processive movement of UNC-104. Our results suggest a common activation mechanism for kinesin-3 family members, while also highlighting their diversification.
Collapse
Affiliation(s)
- Tomoki Kita
- Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
| | - Kyoko Chiba
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku UniversitySendaiJapan
| | - Jiye Wang
- Institute for Protein Research, Osaka UniversityOsakaJapan
| | | | - Shinsuke Niwa
- Graduate School of Life Sciences, Tohoku UniversitySendaiJapan
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku UniversitySendaiJapan
| |
Collapse
|
33
|
Huang J, Korsunsky A, Yazdani M, Chen J. Targeting TRP channels: recent advances in structure, ligand binding, and molecular mechanisms. Front Mol Neurosci 2024; 16:1334370. [PMID: 38273937 PMCID: PMC10808746 DOI: 10.3389/fnmol.2023.1334370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Transient receptor potential (TRP) channels are a large and diverse family of transmembrane ion channels that are widely expressed, have important physiological roles, and are associated with many human diseases. These proteins are actively pursued as promising drug targets, benefitting greatly from advances in structural and mechanistic studies of TRP channels. At the same time, the complex, polymodal activation and regulation of TRP channels have presented formidable challenges. In this short review, we summarize recent progresses toward understanding the structural basis of TRP channel function, as well as potential ligand binding sites that could be targeted for therapeutics. A particular focus is on the current understanding of the molecular mechanisms of TRP channel activation and regulation, where many fundamental questions remain unanswered. We believe that a deeper understanding of the functional mechanisms of TRP channels will be critical and likely transformative toward developing successful therapeutic strategies targeting these exciting proteins. This endeavor will require concerted efforts from computation, structural biology, medicinal chemistry, electrophysiology, pharmacology, drug safety and clinical studies.
Collapse
Affiliation(s)
- Jian Huang
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Aron Korsunsky
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| | - Mahdieh Yazdani
- Modeling and Informatics, Merck & Co., Inc., West Point, PA, United States
| | - Jianhan Chen
- Department of Chemistry, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
34
|
Zheng W, Ziemssen F, Suesskind D, Voykov B, Schnichels S. TRPP2 is located in the primary cilia of human non-pigmented ciliary epithelial cells. Graefes Arch Clin Exp Ophthalmol 2024; 262:93-102. [PMID: 37378878 PMCID: PMC10806040 DOI: 10.1007/s00417-023-06150-w] [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: 01/15/2023] [Revised: 05/30/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
PURPOSE Mechanosensitive channels (MSCs) and primary cilium possess a possible relevance for the sensation of intraocular pressure (IOP). However, there is only limited data on their expression and localization in the ciliary body epithelium (CBE). The purpose of this study was to characterize the expression and localization of TRPP2 in a human non-pigmented ciliary epithelial cell (HNPCE) line. METHODS The expression of the TRPP2 was studied by quantitative (q)RT-PCR and in situ hybridization in rat and human tissue. Protein expression and distribution were studied by western blot analysis, immunohistochemistry, and immunoelectron microscopy. Cellular location of TRPP2 was determined in rat and human CBE by immunofluorescence and immunoblot analysis. Electron microscopy studies were conducted to evaluate where and with substructure TRPP2 is localized in the HNPCE cell line. RESULTS The expression of TRPP2 in rat and human non-pigmented ciliary epithelium was detected. TRPP2 was mainly located in nuclei, but also showed a punctate distribution pattern in the cytoplasm of HNPCE of the tissue and the cell line. In HNPCE cell culture, primary cilia did exhibit different length following serum starvation and hydrostatic pressure. TRPP2 was found to be colocalized with these cilia in HNPCE cells. CONCLUSION The expression of TRPP2 and the primary cilium in the CB may indicate a possible role, such as the sensing of hydrostatic pressure, for the regulation of IOP. Functional studies via patch clamp or pharmacological intervention have yet to clarify the relevance for the physiological situation or aqueous humor regulation.
Collapse
Affiliation(s)
- Wenxu Zheng
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Focke Ziemssen
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany.
- University Eye Hospital Leipzig, Leipzig, Germany.
- Klinik und Poliklinik für Augenheilkunde, Liebigstr. 10-14, 72072, Leipzig, Germany.
| | - Daniela Suesskind
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Bogomil Voykov
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Sven Schnichels
- Centre for Ophthalmology, Eberhard Karls University Tübingen, Tübingen, Germany
| |
Collapse
|
35
|
Bannell TAK, Cockburn JJB. The molecular structure and function of fibrocystin, the key gene product implicated in autosomal recessive polycystic kidney disease (ARPKD). Ann Hum Genet 2024; 88:58-75. [PMID: 37905714 DOI: 10.1111/ahg.12535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/14/2023] [Accepted: 10/03/2023] [Indexed: 11/02/2023]
Abstract
Autosomal recessive polycystic kidney disease is an early onset inherited hepatorenal disorder affecting around 1 in 20,000 births with no approved specific therapies. The disease is almost always caused by variations in the polycystic kidney and hepatic disease 1 gene, which encodes fibrocystin (FC), a very large, single-pass transmembrane glycoprotein found in primary cilia, urine and urinary exosomes. By comparison to proteins involved in autosomal dominant PKD, our structural and molecular understanding of FC has lagged far behind such that there are no published experimentally determined structures of any part of the protein. Bioinformatics analyses predict that the ectodomain contains a long chain of immunoglobulin-like plexin-transcription factor domains, a protective antigen 14 domain, a tandem G8-TMEM2 homology region and a sperm protein, enterokinase and agrin domain. Here we review current knowledge on the molecular function of the protein from a structural perspective.
Collapse
Affiliation(s)
- Travis A K Bannell
- Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Joseph J B Cockburn
- Astbury Centre for Structural and Molecular Biology, University of Leeds, Leeds, UK
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
36
|
Weisser I, Eckberg K, D'Amico S, Buttram D, Aboudehen K. Ablation of Long Noncoding RNA Hoxb3os Exacerbates Cystogenesis in Mouse Polycystic Kidney Disease. J Am Soc Nephrol 2024; 35:41-55. [PMID: 37953472 PMCID: PMC10786614 DOI: 10.1681/asn.0000000000000265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/20/2023] [Indexed: 11/14/2023] Open
Abstract
SIGNIFICANCE STATEMENT Long noncoding RNAs (lncRNAs) are a class of nonprotein coding RNAs with pivotal functions in development and disease. They have emerged as an exciting new drug target category for many common conditions. However, the role of lncRNAs in autosomal dominant polycystic kidney disease (ADPKD) has been understudied. This study provides evidence implicating a lncRNA in the pathogenesis of ADPKD. We report that Hoxb3os is downregulated in ADPKD and regulates mammalian target of rapamycin (mTOR)/Akt pathway in the in vivo mouse kidney. Ablating the expression of Hoxb3os in mouse polycystic kidney disease (PKD) activated mTOR complex 2 (mTORC2) signaling and exacerbated the cystic phenotype. The results from our study provide genetic proof of concept for future studies that focus on targeting lncRNAs as a treatment option in PKD. BACKGROUND ADPKD is a monogenic disorder characterized by the formation of kidney cysts and is primarily caused by mutations in two genes, PKD1 and PKD2 . METHODS In this study, we investigated the role of lncRNA Hoxb3os in ADPKD by ablating its expression in the mouse. RESULTS Hoxb3os -null mice were viable and had grossly normal kidney morphology but displayed activation of mTOR/Akt signaling and subsequent increase in kidney cell proliferation. To determine the role of Hoxb3os in cystogenesis, we crossed the Hoxb3os -null mouse to two orthologous Pkd1 mouse models: Pkhd1/Cre; Pkd1F/F (rapid cyst progression) and Pkd1RC/RC (slow cyst progression). Ablation of Hoxb3os exacerbated cyst growth in both models. To gain insight into the mechanism whereby Hoxb3os inhibition promotes cystogenesis, we performed western blot analysis of mTOR/Akt pathway between Pkd1 single-knockout and Pkd1 - Hoxb3os double-knockout (DKO) mice. Compared with single-knockout, DKO mice presented with enhanced levels of total and phosphorylated Rictor. This was accompanied by increased phosphorylation of Akt at Ser 473 , a known mTORC2 effector site. Physiologically, kidneys from DKO mice displayed between 50% and 60% increase in cell proliferation and cyst number. CONCLUSIONS The results from this study indicate that ablation of Hoxb3os in mouse PKD exacerbates cystogenesis and dysregulates mTORC2.
Collapse
Affiliation(s)
- Ivan Weisser
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Kara Eckberg
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Stephen D'Amico
- Department of Medicine, Stony Brook University, Stony Brook, New York
| | - Daniel Buttram
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Karam Aboudehen
- Department of Medicine, Stony Brook University, Stony Brook, New York
| |
Collapse
|
37
|
Quaglia A, Roberts EA, Torbenson M. Developmental and Inherited Liver Disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:122-294. [DOI: 10.1016/b978-0-7020-8228-3.00003-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
38
|
Kanhai AA, Sánchez-López E, Kuipers TB, van Klinken JB, Dijkstra KL, van der Veen I, Baelde HJ, Song X, Pei Y, Mei H, Leonhard WN, Mayboroda OA, Peters DJ. Short salsalate administration affects cell proliferation, metabolism, and inflammation in polycystic kidney disease. iScience 2023; 26:108278. [PMID: 38026227 PMCID: PMC10665819 DOI: 10.1016/j.isci.2023.108278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/04/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Metabolic reprogramming is a driver of autosomal dominant polycystic kidney disease (ADPKD) progression and a potential therapeutic intervention route. We showed before that the AMP-associated protein kinase (AMPK) activator salsalate attenuates cystic disease progression. Here, we aim to study the early, direct effects of short salsalate treatment in adult-onset conditional Pkd1 deletion mice. Cystic mice were treated with salsalate for two weeks, after which NMR metabolomics and RNA sequencing analyses were performed. Pkd1 deletion resulted in clear metabolomic dysregulation. Short salsalate treatment has small, but significant, effects, reverting acetylcarnitine and phosphocholine concentrations back to wildtype levels, and showing associations with altered purine metabolism. RNA sequencing revealed that short salsalate treatment, next to restoring energy metabolism toward wildtype levels, also affects cell proliferation and inflammation, in PKD. We show that salsalate positively affects major dysregulated processes in ADPKD: energy metabolism, cell proliferation, and inflammation, providing more insights into its working mechanisms.
Collapse
Affiliation(s)
- Anish A. Kanhai
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Elena Sánchez-López
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Thomas B. Kuipers
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Jan B. van Klinken
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- Laboratory Genetic Metabolic Diseases of Pathology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Kyra L. Dijkstra
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Inge van der Veen
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Hans J. Baelde
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Xuewen Song
- Division of Nephrology, University Health Network and University of Toronto, Toronto, ON, Canada
| | - York Pei
- Division of Nephrology, University Health Network and University of Toronto, Toronto, ON, Canada
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Center, Leiden, the Netherlands
| | - Wouter N. Leonhard
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Oleg A. Mayboroda
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, the Netherlands
| | - Dorien J.M. Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
39
|
Jung HJ, Dixon EE, Coleman R, Watnick T, Reiter JF, Outeda P, Cebotaru V, Woodward OM, Welling PA. Polycystin-2-dependent transcriptome reveals early response of autosomal dominant polycystic kidney disease. Physiol Genomics 2023; 55:565-577. [PMID: 37720991 PMCID: PMC11178268 DOI: 10.1152/physiolgenomics.00040.2023] [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: 05/15/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/19/2023] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is caused by mutations in polycystin genes, Pkd1 and Pkd2, but the underlying pathogenic mechanisms are poorly understood. To identify genes and pathways that operate downstream of polycystin-2 (PC2), a comprehensive gene expression database was created, cataloging changes in the transcriptome immediately following PC2 protein depletion. To explore cyst initiation processes, an immortalized mouse inner medullary collecting duct line was developed with the ability to knock out the Pkd2 gene conditionally. Genome-wide transcriptome profiling was performed using RNA sequencing in the cells immediately after PC2 was depleted and compared with isogenic control cells. Differentially expressed genes were identified, and a bioinformatic analysis pipeline was implemented. Altered expression of candidate cystogenic genes was validated in Pkd2 knockout mice. The expression of nearly 900 genes changed upon PC2 depletion. Differentially expressed genes were enriched for genes encoding components of the primary cilia, the canonical Wnt pathway, and MAPK signaling. Among the PC2-dependent ciliary genes, the transcription factor Glis3 was significantly downregulated. MAPK signaling formed a key node at the epicenter of PC2-dependent signaling networks. Activation of Wnt and MAPK signaling, concomitant with the downregulation of Glis3, was corroborated in Pkd2 knockout mice. The data identify a PC2 cilia-to-nucleus signaling axis and dysregulation of the Gli-similar subfamily of transcription factors as a potential initiator of cyst formation in ADPKD. The catalog of PC2-regulated genes should provide a valuable resource for future ADPKD research and new opportunities for drug development.NEW & NOTEWORTHY Autosomal dominant polycystic kidney disease (ADPKD) is the most common inherited kidney disease. Mutations in polycystin genes cause the disease, but the underlying mechanisms of cystogenesis are unknown. To help fill this knowledge gap, we created an inducible cell model of ADPKD and assembled a catalog of genes that respond in immediate proximity to polycystin-2 depletion using transcriptomic profiling. The catalog unveils a ciliary signaling-to-nucleus axis proximal to polycystin-2 dysfunction, highlighting Glis, Wnt, and MAPK signaling.
Collapse
Affiliation(s)
- Hyun Jun Jung
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Eryn E Dixon
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Richard Coleman
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Terry Watnick
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, University of California, San Francisco, California, United States
- Chan Zuckerberg Biohub, San Francisco, California, United States
| | - Patricia Outeda
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Valeriu Cebotaru
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Owen M Woodward
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, United States
| | - Paul A Welling
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| |
Collapse
|
40
|
Koyano T, Fujimoto T, Onishi K, Matsuyama M, Fukushima M, Kume K. Pkd2, mutations linking to autosomal dominant polycystic kidney disease, localizes to the endoplasmic reticulum and regulates calcium signaling in fission yeast. Genes Cells 2023; 28:811-820. [PMID: 37723847 DOI: 10.1111/gtc.13069] [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: 06/30/2023] [Revised: 09/05/2023] [Accepted: 09/07/2023] [Indexed: 09/20/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a renal disorder caused by mutations in the PKD2 gene, which encodes polycystin-2/Pkd2, a transient receptor potential channel. The precise role of Pkd2 in cyst formation remains unclear. The fission yeast Schizosaccharomyces pombe has a putative transient receptor potential channel, Pkd2, which shares similarities with human Pkd2. In this study, truncation analyses of fission yeast Pkd2 were conducted to investigate its localization and function. The results revealed that Pkd2 localizes not only to the plasma membrane but also to the endoplasmic reticulum (ER) in fission yeast. Furthermore, Pkd2 regulates calcium signaling in fission yeast, with the transmembrane domains of Pkd2 being sufficient for these processes. Specifically, the C-terminal region of Pkd2 plays a crucial role in the regulation of calcium signaling. Interestingly, human Pkd2 also localized to the ER and had some impact on calcium signaling in fission yeast. However, human Pkd2 failed to suppress the loss of fission yeast Pkd2. These findings indicate that hPkd2 may not completely substitute for cellular physiology of fission yeast Pkd2. This study provides insights into the localization and functional characteristics of Pkd2 in fission yeast, contributing to our understanding of the pathogenesis of ADPKD.
Collapse
Affiliation(s)
- Takayuki Koyano
- Division of Cell Biology, Shigei Medical Research Institute, Minami-ku, Okayama, Japan
| | - Takahiro Fujimoto
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| | - Kaori Onishi
- Division of Cell Biology, Shigei Medical Research Institute, Minami-ku, Okayama, Japan
| | - Makoto Matsuyama
- Division of Molecular Genetics, Shigei Medical Research Institute, Minami-ku, Okayama, Japan
| | - Masaki Fukushima
- Division of Molecular Genetics, Shigei Medical Research Institute, Minami-ku, Okayama, Japan
- Shigei Medical Research Hospital, Minami-ku, Okayama, Japan
| | - Kazunori Kume
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
- Hiroshima Research Center for Healthy Aging (HiHA), Hiroshima University, Higashi-Hiroshima, Hiroshima, Japan
| |
Collapse
|
41
|
Pala R, Barui AK, Mohieldin AM, Zhou J, Nauli SM. Folate conjugated nanomedicines for selective inhibition of mTOR signaling in polycystic kidneys at clinically relevant doses. Biomaterials 2023; 302:122329. [PMID: 37722182 PMCID: PMC10836200 DOI: 10.1016/j.biomaterials.2023.122329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Although rapamycin is a very effective drug for rodents with polycystic kidney disease (PKD), it is not encouraging in the clinical trials due to the suboptimal dosages compelled by the off-target side effects. We here report the generation, characterization, specificity, functionality, pharmacokinetic, pharmacodynamic and toxicology profiles of novel polycystic kidney-specific-targeting nanoparticles (NPs). We formulated folate-conjugated PLGA-PEG NPs, which can be loaded with multiple drugs, including rapamycin (an mTOR inhibitor) and antioxidant 4-hydroxy-TEMPO (a nephroprotective agent). The NPs increased the efficacy, potency and tolerability of rapamycin resulting in an increased survival rate and improved kidney function by decreasing side effects and reducing biodistribution to other organs in PKD mice. The daily administration of rapamycin-alone (1 mg/kg/day) could now be achieved with a weekly injection of NPs containing rapamycin (379 μg/kg/week). This polycystic kidney-targeting nanotechnology, for the first time, integrated advances in the use of 1) nanoparticles as a delivery cargo, 2) folate for targeting, 3) near-infrared Cy5-fluorophore for in vitro and in vivo live imaging, 4) rapamycin as a pharmacological therapy, and 5) TEMPO as a combinational therapy. The slow sustained-release of rapamycin by polycystic kidney-targeting NPs demonstrates a new era of nanomedicine in treatment for chronic kidney diseases at clinically relevant doses.
Collapse
Affiliation(s)
- Rajasekharreddy Pala
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, 92618, USA; Marlin Biopharma, Irvine, CA, 92620, USA.
| | - Ayan K Barui
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, 92618, USA
| | - Ashraf M Mohieldin
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, 92618, USA
| | - Jing Zhou
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Chapman University, Irvine, CA, 92618, USA; Marlin Biopharma, Irvine, CA, 92620, USA.
| |
Collapse
|
42
|
Trutin I, Oletić L, Nikuševa-Martić T. Unraveling the Complexity of Childhood Polycystic Kidney Disease: A Case Study of Three Sisters. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1700. [PMID: 37892363 PMCID: PMC10605507 DOI: 10.3390/children10101700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/11/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common hereditary kidney disorder, estimated to affect 1 in 1000 people. It displays a high level of variability in terms of onset and severity among affected individuals within the same family. In this case study, three sisters (4, 8, and 10 years of age) were suspected of having ADPKD due to their positive family history. While the two younger sisters aged 8 and 4 showed no disease complications and had normal kidney function, the oldest sister was found to have no dipping status on ambulatory blood pressure measurement (ABPM). Two of the sisters were discovered to have a PKD1 mutation, while the third sister aged 8 was heterozygous for TTC21B c.1593_1595del, p. (Leu532del), which is a variant of uncertain significance (VUS). Environmental factors and genetic modifying factors are believed to contribute to the phenotypic variability observed in ADPKD. Identifying and understanding potential genetic and environmental modifiers of ADPKD could pave the way to targeted treatments for childhood ADPKD.
Collapse
Affiliation(s)
- Ivana Trutin
- Department of Pediatrics, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (I.T.); (L.O.)
| | - Lea Oletić
- Department of Pediatrics, Sestre Milosrdnice University Hospital Center, 10000 Zagreb, Croatia; (I.T.); (L.O.)
| | | |
Collapse
|
43
|
Romano S, Marcon D, Branz L, Tagetti A, Monamì G, Giontella A, Malesani F, Pecoraro L, Minuz P, Brugnara M, Fava C. Subclinical Target Organ Damage in a Sample of Children with Autosomal Dominant Polycystic Kidney Disease: A Pilot Study. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1777. [PMID: 37893495 PMCID: PMC10608453 DOI: 10.3390/medicina59101777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 09/21/2023] [Accepted: 10/02/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Hypertension and vascular damage can begin in adolescents affected by Autosomal Dominant Polycystic Kidney Disease (ADPKD). This study aimed to evaluate markers of vascular damage and left ventricular geometry in a sample of children with ADPKD. Materials and Methods: Several vascular measurements were obtained: ambulatory blood pressure monitoring (ABPM), carotid intima-media thickness (cIMT), carotid distensibility coefficient (cDC), pulse wave velocity (PWV), and echocardiographic measurements (relative wall thickness (RWT) and left ventricular mass index (LVMI)). Results: Eleven ADPKD children were recruited (four females and seven males, mean age 9.5 ± 3.2 years). Four children were hypertensive at the ABPM, five were normotensive, and for two ABPM was not available. RWT was tendentially high (mean 0.47 ± 0.39). Eight patients had concentric cardiac remodeling, while one patient had cardiac hypertrophy. cIMT was above the 95° percentile for sex and height in 80% of the children (0.5 ± 0.005 mm). The average PWV and cDC were between the normal range (5.5 ± 4.6 m/s and 89.6 ± 16.1 × 10-3/KPa, respectively). We observed a positive correlation between the PWV and RWT (r = 0.616; p = 0.044) and a negative correlation between cDC and RWT (r = -0.770; p = 0.015). Cardiovascular damages (cIMT > 95° percentile) were found in normotensive patients. Conclusions: Increased RWT and high cIMT, indicating subclinical organ damage, are already present in ADPKD children. RWT was significantly correlated to that of cDC and PWV, implying that vascular stiffening is associated with cardiac remodeling. None of the children had an alteration in renal function. Subclinical cardiovascular damage preceded the decline in glomerular filtration rate.
Collapse
Affiliation(s)
- Simone Romano
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Denise Marcon
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Lorella Branz
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Angela Tagetti
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Giada Monamì
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Alice Giontella
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Francesca Malesani
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Luca Pecoraro
- Pediatric Clinic, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy
| | - Pietro Minuz
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| | - Milena Brugnara
- Pediatric Clinic, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, 37126 Verona, Italy
| | - Cristiano Fava
- Section of General Medicine and Hypertension, Department of Medicine, Policlinico GB Rossi, University of Verona, 37134 Verona, Italy (D.M.); (L.B.); (A.G.); (P.M.); (C.F.)
| |
Collapse
|
44
|
Kataoka H, Shimada Y, Kimura T, Nishio S, Nakatani S, Mochizuki T, Tsuchiya K, Hoshino J, Hattanda F, Kawano H, Hanaoka K, Hidaka S, Ichikawa D, Ishikawa E, Uchiyama K, Hayashi H, Makabe S, Manabe S, Mitobe M, Sekine A, Suwabe T, Kai H, Kurashige M, Seta K, Shimazu K, Moriyama T, Sato M, Otsuka T, Katayama K, Shimabukuro W, Fujimaru T, Miura K, Nakanishi K, Horie S, Furuichi K, Okada H, Narita I, Muto S. Public support for patients with intractable diseases in Japan: impact on clinical indicators from nationwide registries in patients with autosomal dominant polycystic kidney disease. Clin Exp Nephrol 2023; 27:809-818. [PMID: 37368094 DOI: 10.1007/s10157-023-02372-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 06/11/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Clinical practice guidelines recommend antihypertensive and tolvaptan therapies for patients with autosomal dominant polycystic kidney disease (ADPKD) in Japan. However, tolvaptan therapy may pose an economic burden. The Japanese Ministry of Health, Labour and Welfare supports patients with intractable diseases. This study aimed to confirm the impact of the intractable disease system in Japan on the clinical treatment of ADPKD. METHODS We analyzed the data of 3768 patients with ADPKD having a medical subsidy certificate from the Japanese Ministry of Health, Labour and Welfare in 2015-2016. The following quality indicators were use: the adherence rate to the 2014 clinical practice guideline for polycystic kidney disease (prescription rates of antihypertensive agents and tolvaptan in this cohort) and the number of Japanese patients with ADPKD nationwide started on renal replacement therapy in 2014 and 2020. RESULTS Compared with new applications from 2015 to 2016, the prescription rates of antihypertensives and tolvaptan for the indicated patients at the 2017 renewal application increased by 2.0% (odds ratio = 1.41, p = 0.008) and 47.4% (odds ratio = 10.1, p > 0.001), respectively. These quality indicators improved with antihypertensive treatment, especially in patients with chronic kidney disease stages 1-2 (odds ratio = 1.79, p = 0.013) and in those aged < 50 years (odds ratio = 1.70, p = 0.003). The number of patients with ADPKD who were started on renal replacement therapy in Japan decreased from 999 in 2014 to 884 in 2020 in the nationwide database (odds ratio = 0.83, p < 0.001). CONCLUSIONS The Japanese public intractable disease support system contributes to improvement of ADPKD treatment.
Collapse
Affiliation(s)
- Hiroshi Kataoka
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Yosuke Shimada
- Intelligent Systems Laboratory, SECOM CO., LTD. Mitaka, Tokyo, Japan
- Department of Medical Eleactronic Intelligence Management, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomonori Kimura
- Reverse Translational Research Project, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Ibaraki, Osaka, Japan
| | - Saori Nishio
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Nakatani
- Department of Metabolism, Endocrinology and Molecular Medicine, Osaka City University Graduate School of Medicine Osaka, Osaka, Japan
| | - Toshio Mochizuki
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
- PKD Nephrology Clinic, Tokyo, Japan
| | - Ken Tsuchiya
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Junichi Hoshino
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Fumihiko Hattanda
- Department of Rheumatology, Endocrinology and Nephrology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Haruna Kawano
- Department of Urology, Department of Advanced Informatics for Genetic Disease, Juntendo University, Tokyo, Japan
| | - Kazushige Hanaoka
- Department of General Internal Medicine, The Jikei University, Tokyo, Japan
| | - Sumi Hidaka
- Kidney Disease and Transplant Center, Shonan Kamakura General Hospital, Kanagawa, Japan
| | - Daisuke Ichikawa
- Department of Nephrology and Hypertension, St Marianna University School of Medicine, Kanagawa, Japan
| | - Eiji Ishikawa
- Department of Internal Medicine, Saiseikai Matsusaka General Hospital, Mie, Japan
| | - Kiyotaka Uchiyama
- Division of Endocrinology, Metabolism and Nephrology Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Hiroki Hayashi
- Department of Nephrology, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Shiho Makabe
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Shun Manabe
- Department of Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Michihiro Mitobe
- Department of Nephrology, Takeda General Hospital, Fukushima, Japan
| | - Akinari Sekine
- Department of Nephrology, Toranomon Hospital, Tokyo, Japan
| | - Tatsuya Suwabe
- Department of Nephrology, Toranomon Hospital, Tokyo, Japan
| | - Hirayasu Kai
- Pathophysiology of Renal Diseases, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Mahiro Kurashige
- Nephrology and Hypertension, Department of Internal Medicine, The Jikei University, Tokyo, Japan
| | - Koichi Seta
- Department of Nephrology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Keiji Shimazu
- Department of Nephrology, Osaka Saiseikai Nakatsu Hospital, Osaka, Japan
| | - Tomofumi Moriyama
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Fukuoka, Japan
| | - Mai Sato
- Division of Nephrology and Rheumatology, National Center for Child Health and Development, Tokyo, Japan
| | - Tadashi Otsuka
- Division of Clinical Nephrology and Rheumatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Kan Katayama
- Department of Cardiology and Nephrology, Mie University Graduate School of Medicine, Mie, Japan
| | - Wataru Shimabukuro
- Department of Child Health and Welfare (Pediatrics), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Takuya Fujimaru
- Department of Nephrology, St. Luke's International Hospital, 9-1 Akashi-Cho, Chuo-Ku, Tokyo, Japan
| | - Kenichiro Miura
- Department of Pediatric Nephrology, Tokyo Women's Medical University, Tokyo, Japan
| | - Koichi Nakanishi
- Department of Child Health and Welfare (Pediatrics), Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan
| | - Shigeo Horie
- Department of Urology, Department of Advanced Informatics for Genetic Disease, Juntendo University, Tokyo, Japan
| | - Kengo Furuichi
- Department of Nephrology, Kanazawa Medical University, Ishikawa, Japan
| | - Hirokazu Okada
- Department of Nephrology, Saitama Medical University, Saitama, Japan
| | - Ichiei Narita
- Division of Clinical Nephrology and Rheumatology, Niigata University, Niigata, Japan
| | - Satoru Muto
- Department of Urology, Juntendo University Nerima Hospital, Tokyo, Japan.
| |
Collapse
|
45
|
Saito M, Otsu W, Miyadera K, Nishimura Y. Recent advances in the understanding of cilia mechanisms and their applications as therapeutic targets. Front Mol Biosci 2023; 10:1232188. [PMID: 37780208 PMCID: PMC10538646 DOI: 10.3389/fmolb.2023.1232188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 08/24/2023] [Indexed: 10/03/2023] Open
Abstract
The primary cilium is a single immotile microtubule-based organelle that protrudes into the extracellular space. Malformations and dysfunctions of the cilia have been associated with various forms of syndromic and non-syndromic diseases, termed ciliopathies. The primary cilium is therefore gaining attention due to its potential as a therapeutic target. In this review, we examine ciliary receptors, ciliogenesis, and ciliary trafficking as possible therapeutic targets. We first discuss the mechanisms of selective distribution, signal transduction, and physiological roles of ciliary receptors. Next, pathways that regulate ciliogenesis, specifically the Aurora A kinase, mammalian target of rapamycin, and ubiquitin-proteasome pathways are examined as therapeutic targets to regulate ciliogenesis. Then, in the photoreceptors, the mechanism of ciliary trafficking which takes place at the transition zone involving the ciliary membrane proteins is reviewed. Finally, some of the current therapeutic advancements highlighting the role of large animal models of photoreceptor ciliopathy are discussed.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Physiology and Pathology, School of Pharma-Sciences, Teikyo University, Tokyo, Japan
| | - Wataru Otsu
- Department of Biomedical Research Laboratory, Gifu Pharmaceutical University, Gifu, Japan
| | - Keiko Miyadera
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Yuhei Nishimura
- Department of Integrative Pharmacology, Mie University Graduate School of Medicine, Tsu, Mie, Japan
- Mie University Research Center for Cilia and Diseases, Tsu, Mie, Japan
| |
Collapse
|
46
|
Peng C, Chen H, Ren J, Zhou F, Li Y, Keqie Y, Ding T, Ruan J, Wang H, Chen X, Liu S. A long-read sequencing and SNP haplotype-based novel preimplantation genetic testing method for female ADPKD patient with de novo PKD1 mutation. BMC Genomics 2023; 24:521. [PMID: 37667185 PMCID: PMC10478289 DOI: 10.1186/s12864-023-09593-x] [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: 01/16/2023] [Accepted: 08/16/2023] [Indexed: 09/06/2023] Open
Abstract
The autosomal dominant form of polycystic kidney disease (ADPKD) is the most common hereditary disease that causes late-onset renal cyst development and end-stage renal disease. Preimplantation genetic testing for monogenic disease (PGT-M) has emerged as an effective strategy to prevent pathogenic mutation transmission rely on SNP linkage analysis between pedigree members. Yet, it remains challenging to establish reliable PGT-M methods for ADPKD cases or other monogenic diseases with de novo mutations or without a family history. Here we reported the application of long-read sequencing for direct haplotyping in a female patient with de novo PKD1 c.11,526 G > C mutation and successfully established the high-risk haplotype. Together with targeted short-read sequencing of SNPs for the couple and embryos, the carrier status for embryos was identified. A healthy baby was born without the PKD1 pathogenic mutation. Our PGT-M strategy based on long-read sequencing for direct haplotyping combined with targeted SNP haplotype can be widely applied to other monogenic disease carriers with de novo mutation.
Collapse
Affiliation(s)
- Cuiting Peng
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | - Han Chen
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | - Jun Ren
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | - Fan Zhou
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | - Yutong Li
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | - Yuezhi Keqie
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | | | | | - He Wang
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China
| | - Xinlian Chen
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China.
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China.
| | - Shanling Liu
- Center of prenatal diagnosis, Department of Medical Genetics, West China Second University Hospital, Sichuan University, No17, Section 3, South Renmin Road, Chengdu, China.
- Laboratory of birth defects and related diseases of women and children, Sichuan university, Ministry of Education, Sichuan, China.
| |
Collapse
|
47
|
Bu LK, Jia PP, Li WG, Li YZ, Li TY, Pei DS. Probiotics mitigate kidney damage after exposure to Sri Lanka's local groundwater from chronic kidney disease with uncertain etiology (CKDu) prevalent area in zebrafish. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 262:106671. [PMID: 37657145 DOI: 10.1016/j.aquatox.2023.106671] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/03/2023]
Abstract
Groundwater in Sri Lanka, contaminated with environmental toxins, is suspected to potentially induce chronic kidney disease of uncertain etiology (CKDu) in humans. This study aims to elucidate the potential mitigating effects of probiotics on kidney damage induced by exposure to this local groundwater (LW) in zebrafish. We used zebrafish as a model organism and exposed them to local groundwater to evaluate the risk of CKDu. Probiotics were then added at a concentration of 108 colony-forming units per milliliter (CFU/mL). Our findings revealed that exposure to local groundwater resulted in abnormalities, such as tail deletion and spinal curvature in zebrafish larvae. However, the addition of probiotics mitigated these effects, improving the hatching rate, heart rate, length, weight, deformity rate, survival rate, and abnormal behavior of zebrafish. It also positively influenced the differential expression levels of kidney development and immunity-related genes (dync2h1, foxj1, pkd2, gata3, slc20a1, il1β, and lyso). Furthermore, exposure to LW decreased both the diversity and abundance of microbiota in zebrafish larvae. However, treatment with probiotics, such as L. plantarum and L. rhamnosus partially restored the disrupted gut microbiota and significantly impacted the cellular process pathways of the microbial community, as determined by KEGG (Kyoto Encyclopedia of Genes and Genomes) analysis. In conclusion, this study highlights the risks associated with Sri Lanka's local groundwater from a CKDu prevalent area and confirms the beneficial effects of different probiotics. These findings may provide new insights into bacterial function in host kidney health.
Collapse
Affiliation(s)
- Ling-Kang Bu
- College of Life Science, Henan Normal University, Xinxiang 453007, China; School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Pan-Pan Jia
- School of Public Health, Chongqing Medical University, Chongqing 400016, China
| | - Wei-Guo Li
- College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Yong-Zhi Li
- Chongqing Institute of Green and Intelligent Technology, Chongqing School of University of Chinese Academy of Sciences, CAS, Chongqing 400714, China
| | - Tian-Yun Li
- Chongqing Institute of Green and Intelligent Technology, Chongqing School of University of Chinese Academy of Sciences, CAS, Chongqing 400714, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
48
|
Derderian C, Canales GI, Reiter JF. Seriously cilia: A tiny organelle illuminates evolution, disease, and intercellular communication. Dev Cell 2023; 58:1333-1349. [PMID: 37490910 PMCID: PMC10880727 DOI: 10.1016/j.devcel.2023.06.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/18/2023] [Accepted: 06/30/2023] [Indexed: 07/27/2023]
Abstract
The borders between cell and developmental biology, which have always been permeable, have largely dissolved. One manifestation is the blossoming of cilia biology, with cell and developmental approaches (increasingly complemented by human genetics, structural insights, and computational analysis) fruitfully advancing understanding of this fascinating, multifunctional organelle. The last eukaryotic common ancestor probably possessed a motile cilium, providing evolution with ample opportunity to adapt cilia to many jobs. Over the last decades, we have learned how non-motile, primary cilia play important roles in intercellular communication. Reflecting their diverse motility and signaling functions, compromised cilia cause a diverse range of diseases collectively called "ciliopathies." In this review, we highlight how cilia signal, focusing on how second messengers generated in cilia convey distinct information; how cilia are a potential source of signals to other cells; how evolution may have shaped ciliary function; and how cilia research may address thorny outstanding questions.
Collapse
Affiliation(s)
- Camille Derderian
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriela I Canales
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jeremy F Reiter
- Department of Biochemistry and Biophysics, Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
49
|
Sieben CJ, Harris PC. Experimental Models of Polycystic Kidney Disease: Applications and Therapeutic Testing. KIDNEY360 2023; 4:1155-1173. [PMID: 37418622 PMCID: PMC10476690 DOI: 10.34067/kid.0000000000000209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 06/29/2023] [Indexed: 07/09/2023]
Abstract
Polycystic kidney diseases (PKDs) are genetic disorders characterized by the formation and expansion of numerous fluid-filled renal cysts, damaging normal parenchyma and often leading to kidney failure. Although PKDs comprise a broad range of different diseases, with substantial genetic and phenotypic heterogeneity, an association with primary cilia represents a common theme. Great strides have been made in the identification of causative genes, furthering our understanding of the genetic complexity and disease mechanisms, but only one therapy so far has shown success in clinical trials and advanced to US Food and Drug Administration approval. A key step in understanding disease pathogenesis and testing potential therapeutics is developing orthologous experimental models that accurately recapitulate the human phenotype. This has been particularly important for PKDs because cellular models have been of limited value; however, the advent of organoid usage has expanded capabilities in this area but does not negate the need for whole-organism models where renal function can be assessed. Animal model generation is further complicated in the most common disease type, autosomal dominant PKD, by homozygous lethality and a very limited cystic phenotype in heterozygotes while for autosomal recessive PKD, mouse models have a delayed and modest kidney disease, in contrast to humans. However, for autosomal dominant PKD, the use of conditional/inducible and dosage models have resulted in some of the best disease models in nephrology. These have been used to help understand pathogenesis, to facilitate genetic interaction studies, and to perform preclinical testing. Whereas for autosomal recessive PKD, using alternative species and digenic models has partially overcome these deficiencies. Here, we review the experimental models that are currently available and most valuable for therapeutic testing in PKD, their applications, success in preclinical trials, advantages and limitations, and where further improvements are needed.
Collapse
Affiliation(s)
- Cynthia J Sieben
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | | |
Collapse
|
50
|
Zhang M, Ma Y, Ye X, Zhang N, Pan L, Wang B. TRP (transient receptor potential) ion channel family: structures, biological functions and therapeutic interventions for diseases. Signal Transduct Target Ther 2023; 8:261. [PMID: 37402746 DOI: 10.1038/s41392-023-01464-x] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/26/2023] [Accepted: 04/25/2023] [Indexed: 07/06/2023] Open
Abstract
Transient receptor potential (TRP) channels are sensors for a variety of cellular and environmental signals. Mammals express a total of 28 different TRP channel proteins, which can be divided into seven subfamilies based on amino acid sequence homology: TRPA (Ankyrin), TRPC (Canonical), TRPM (Melastatin), TRPML (Mucolipin), TRPN (NO-mechano-potential, NOMP), TRPP (Polycystin), TRPV (Vanilloid). They are a class of ion channels found in numerous tissues and cell types and are permeable to a wide range of cations such as Ca2+, Mg2+, Na+, K+, and others. TRP channels are responsible for various sensory responses including heat, cold, pain, stress, vision and taste and can be activated by a number of stimuli. Their predominantly location on the cell surface, their interaction with numerous physiological signaling pathways, and the unique crystal structure of TRP channels make TRPs attractive drug targets and implicate them in the treatment of a wide range of diseases. Here, we review the history of TRP channel discovery, summarize the structures and functions of the TRP ion channel family, and highlight the current understanding of the role of TRP channels in the pathogenesis of human disease. Most importantly, we describe TRP channel-related drug discovery, therapeutic interventions for diseases and the limitations of targeting TRP channels in potential clinical applications.
Collapse
Affiliation(s)
- Miao Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yueming Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xianglu Ye
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ning Zhang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Lei Pan
- The Center for Microbes, Development and Health; Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
- CAS Center for Excellence in Biotic Interactions, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Bing Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
- Center for Pharmaceutics Research, Shanghai Institute of Materia Medica Chinese Academy of Sciences, Shanghai, 201203, China.
| |
Collapse
|