1
|
Zhou JX, Li LX, Zhang H, Agborbesong E, Harris PC, Calvet JP, Li X. DNA methyltransferase 1 (DNMT1) promotes cyst growth and epigenetic age acceleration in autosomal dominant polycystic kidney disease. Kidney Int 2024; 106:258-272. [PMID: 38782200 PMCID: PMC11270650 DOI: 10.1016/j.kint.2024.04.017] [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: 06/15/2023] [Revised: 02/28/2024] [Accepted: 04/05/2024] [Indexed: 05/25/2024]
Abstract
Alteration of DNA methylation leads to diverse diseases, and the dynamic changes of DNA methylation (DNAm) on sets of CpG dinucleotides in mammalian genomes are termed "DNAm age" and "epigenetic clocks" that can predict chronological age. However, whether and how dysregulation of DNA methylation promotes cyst progression and epigenetic age acceleration in autosomal dominant polycystic kidney disease (ADPKD) remains elusive. Here, we show that DNA methyltransferase 1 (DNMT1) is upregulated in cystic kidney epithelial cells and tissues and that knockout of Dnmt1 and targeting DNMT1 with hydralazine, a safe demethylating agent, delays cyst growth in Pkd1 mutant kidneys and extends life span of Pkd1 conditional knockout mice. With methyl-CpG binding domain (MBD) protein-enriched genome sequencing (MBD-seq), DNMT1 chromatin immunoprecipitation (ChIP)-sequencing and RNA-sequencing analysis, we identified two novel DNMT1 targets, PTPRM and PTPN22 (members of the protein tyrosine phosphatase family). PTPRM and PTPN22 function as mediators of DNMT1 and the phosphorylation and activation of PKD-associated signaling pathways, including ERK, mTOR and STAT3. With whole-genome bisulfide sequencing in kidneys of patients with ADPKD versus normal individuals, we found that the methylation of epigenetic clock-associated genes was dysregulated, supporting that epigenetic age is accelerated in the kidneys of patients with ADPKD. Furthermore, five epigenetic clock-associated genes, including Hsd17b14, Itpkb, Mbnl1, Rassf5 and Plk2, were identified. Thus, the diverse biological roles of these five genes suggest that their methylation status may not only predict epigenetic age acceleration but also contribute to disease progression in ADPKD.
Collapse
Affiliation(s)
- Julie Xia Zhou
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA.
| | - Linda Xiaoyan Li
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Hongbing Zhang
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ewud Agborbesong
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Peter C Harris
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - James P Calvet
- Department of Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Xiaogang Li
- Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA.
| |
Collapse
|
2
|
Yu JT, Fan S, Li XY, Hou R, Hu XW, Wang JN, Shan RR, Dong ZH, Xie MM, Dong YH, Shen XY, Jin J, Wen JG, Liu MM, Wang W, Meng XM. Novel insights into STAT3 in renal diseases. Biomed Pharmacother 2023; 165:115166. [PMID: 37473682 DOI: 10.1016/j.biopha.2023.115166] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/22/2023] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a cell-signal transcription factor that has attracted considerable attention in recent years. The stimulation of cytokines and growth factors can result in the transcription of a wide range of genes that are crucial for several cellular biological processes involved in pro- and anti-inflammatory responses. STAT3 has attracted considerable interest as a result of a recent upsurge in study because of their role in directing the innate immune response and sustaining inflammatory pathways, which is a key feature in the pathogenesis of many diseases, including renal disorders. Several pathological conditions which may involve STAT3 include diabetic nephropathy, acute kidney injury, lupus nephritis, polycystic kidney disease, and renal cell carcinoma. STAT3 is expressed in various renal tissues under these pathological conditions. To better understand the role of STAT3 in the kidney and provide a theoretical foundation for STAT3-targeted therapy for renal disorders, this review covers the current work on the activities of STAT3 and its mechanisms in the pathophysiological processes of various types of renal diseases.
Collapse
Affiliation(s)
- Ju-Tao Yu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shuai Fan
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230032 China; Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032 China
| | - Xiang-Yu Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Rui Hou
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Wei Hu
- Department of Clinical Pharmacy, Anhui Provincial Children's Hospital, Hefei 230051, China
| | - Jia-Nan Wang
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Run-Run Shan
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Ze-Hui Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Man-Man Xie
- School of Life Sciences, Anhui Medical University, Hefei 230032, China
| | - Yu-Hang Dong
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xiao-Yu Shen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Juan Jin
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei 230032, China
| | - Jia-Gen Wen
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ming-Ming Liu
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China
| | - Wei Wang
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Hefei 230032 China; Department of Urology, Institute of Urology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032 China.
| | - Xiao-Ming Meng
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, the Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
3
|
Leiding JW, Vogel TP, Santarlas VGJ, Mhaskar R, Smith MR, Carisey A, Vargas-Hernández A, Silva-Carmona M, Heeg M, Rensing-Ehl A, Neven B, Hadjadj J, Hambleton S, Ronan Leahy T, Meesilpavikai K, Cunningham-Rundles C, Dutmer CM, Sharapova SO, Taskinen M, Chua I, Hague R, Klemann C, Kostyuchenko L, Morio T, Thatayatikom A, Ozen A, Scherbina A, Bauer CS, Flanagan SE, Gambineri E, Giovannini-Chami L, Heimall J, Sullivan KE, Allenspach E, Romberg N, Deane SG, Prince BT, Rose MJ, Bohnsack J, Mousallem T, Jesudas R, Santos Vilela MMD, O'Sullivan M, Pachlopnik Schmid J, Průhová Š, Klocperk A, Rees M, Su H, Bahna S, Baris S, Bartnikas LM, Chang Berger A, Briggs TA, Brothers S, Bundy V, Chan AY, Chandrakasan S, Christiansen M, Cole T, Cook MC, Desai MM, Fischer U, Fulcher DA, Gallo S, Gauthier A, Gennery AR, Gonçalo Marques J, Gottrand F, Grimbacher B, Grunebaum E, Haapaniemi E, Hämäläinen S, Heiskanen K, Heiskanen-Kosma T, Hoffman HM, Gonzalez-Granado LI, Guerrerio AL, Kainulainen L, Kumar A, Lawrence MG, Levin C, Martelius T, Neth O, Olbrich P, Palma A, Patel NC, Pozos T, Preece K, Lugo Reyes SO, Russell MA, Schejter Y, Seroogy C, Sinclair J, Skevofilax E, Suan D, Suez D, Szabolcs P, Velasco H, Warnatz K, Walkovich K, et alLeiding JW, Vogel TP, Santarlas VGJ, Mhaskar R, Smith MR, Carisey A, Vargas-Hernández A, Silva-Carmona M, Heeg M, Rensing-Ehl A, Neven B, Hadjadj J, Hambleton S, Ronan Leahy T, Meesilpavikai K, Cunningham-Rundles C, Dutmer CM, Sharapova SO, Taskinen M, Chua I, Hague R, Klemann C, Kostyuchenko L, Morio T, Thatayatikom A, Ozen A, Scherbina A, Bauer CS, Flanagan SE, Gambineri E, Giovannini-Chami L, Heimall J, Sullivan KE, Allenspach E, Romberg N, Deane SG, Prince BT, Rose MJ, Bohnsack J, Mousallem T, Jesudas R, Santos Vilela MMD, O'Sullivan M, Pachlopnik Schmid J, Průhová Š, Klocperk A, Rees M, Su H, Bahna S, Baris S, Bartnikas LM, Chang Berger A, Briggs TA, Brothers S, Bundy V, Chan AY, Chandrakasan S, Christiansen M, Cole T, Cook MC, Desai MM, Fischer U, Fulcher DA, Gallo S, Gauthier A, Gennery AR, Gonçalo Marques J, Gottrand F, Grimbacher B, Grunebaum E, Haapaniemi E, Hämäläinen S, Heiskanen K, Heiskanen-Kosma T, Hoffman HM, Gonzalez-Granado LI, Guerrerio AL, Kainulainen L, Kumar A, Lawrence MG, Levin C, Martelius T, Neth O, Olbrich P, Palma A, Patel NC, Pozos T, Preece K, Lugo Reyes SO, Russell MA, Schejter Y, Seroogy C, Sinclair J, Skevofilax E, Suan D, Suez D, Szabolcs P, Velasco H, Warnatz K, Walkovich K, Worth A, Seppänen MRJ, Torgerson TR, Sogkas G, Ehl S, Tangye SG, Cooper MA, Milner JD, Forbes Satter LR. Monogenic early-onset lymphoproliferation and autoimmunity: Natural history of STAT3 gain-of-function syndrome. J Allergy Clin Immunol 2023; 151:1081-1095. [PMID: 36228738 PMCID: PMC10081938 DOI: 10.1016/j.jaci.2022.09.002] [Show More Authors] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 08/29/2022] [Accepted: 09/01/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUND In 2014, germline signal transducer and activator of transcription (STAT) 3 gain-of-function (GOF) mutations were first described to cause a novel multisystem disease of early-onset lymphoproliferation and autoimmunity. OBJECTIVE This pivotal cohort study defines the scope, natural history, treatment, and overall survival of a large global cohort of patients with pathogenic STAT3 GOF variants. METHODS We identified 191 patients from 33 countries with 72 unique mutations. Inclusion criteria included symptoms of immune dysregulation and a biochemically confirmed germline heterozygous GOF variant in STAT3. RESULTS Overall survival was 88%, median age at onset of symptoms was 2.3 years, and median age at diagnosis was 12 years. Immune dysregulatory features were present in all patients: lymphoproliferation was the most common manifestation (73%); increased frequencies of double-negative (CD4-CD8-) T cells were found in 83% of patients tested. Autoimmune cytopenias were the second most common clinical manifestation (67%), followed by growth delay, enteropathy, skin disease, pulmonary disease, endocrinopathy, arthritis, autoimmune hepatitis, neurologic disease, vasculopathy, renal disease, and malignancy. Infections were reported in 72% of the cohort. A cellular and humoral immunodeficiency was observed in 37% and 51% of patients, respectively. Clinical symptoms dramatically improved in patients treated with JAK inhibitors, while a variety of other immunomodulatory treatment modalities were less efficacious. Thus far, 23 patients have undergone bone marrow transplantation, with a 62% survival rate. CONCLUSION STAT3 GOF patients present with a wide array of immune-mediated disease including lymphoproliferation, autoimmune cytopenias, and multisystem autoimmunity. Patient care tends to be siloed, without a clear treatment strategy. Thus, early identification and prompt treatment implementation are lifesaving for STAT3 GOF syndrome.
Collapse
Affiliation(s)
- Jennifer W Leiding
- Division of Allergy and Immunology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore; Johns Hopkins All Children's Institute for Clinical and Translational Research, Johns Hopkins All Children's Hospital, St Petersburg.
| | - Tiphanie P Vogel
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston
| | | | - Rahul Mhaskar
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa
| | - Madison R Smith
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston
| | - Alexandre Carisey
- Department of Cell and Molecular Biology, St Jude Children's Research Hospital, Memphis
| | - Alexander Vargas-Hernández
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston
| | - Manuel Silva-Carmona
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Anne Rensing-Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Bénédicte Neven
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163-Institut Imagine, Paris
| | - Jérôme Hadjadj
- Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163-Institut Imagine, Paris
| | - Sophie Hambleton
- Newcastle University Translational and Clinical Research Institute, Newcastle (United Kingdom)
| | | | - Kornvalee Meesilpavikai
- Department of Internal Medicine, Division of Clinical Immunology and Department of Immunology, Erasmus University Medical Center, Rotterdam, Netherlands; Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Cullen M Dutmer
- Children's Hospital Colorado, University of Colorado School of Medicine, Aurora
| | - Svetlana O Sharapova
- Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk
| | - Mervi Taskinen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Turku and Kuopio, Finland
| | - Ignatius Chua
- Department of Rheumatology, Immunology and Allergy, Christchurch Hospital, Christchurch; Clinical Immunogenomics Research Consortium of Australasia (CIRCA)
| | | | - Christian Klemann
- Department of Pediatric Pneumology, Allergy and Neonatology, Hannover Medical School, Hannover
| | - Larysa Kostyuchenko
- Center of Pediatric Immunology, Western Ukrainian Specialized Children's Medical Centre, Lviv
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo
| | - Akaluck Thatayatikom
- Division of Pediatric Allergy/Immunology/Rheumatology, Shands Children's Hospital, University of Florida, Gainesville
| | - Ahmet Ozen
- School of Medicine, Pediatric Allergy and Immunology, Marmara University, Istanbul
| | - Anna Scherbina
- Dmitry Rogachev National Medical and Research Center for Pediatric Hematology, Oncology and Immunology, Moscow
| | - Cindy S Bauer
- Division of Allergy and Immunology, Phoenix Children's Hospital, Phoenix
| | - Sarah E Flanagan
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter
| | - Eleonora Gambineri
- Department of NEUROFARBA, Section of Children's Health, University of Florence, Anna Meyer Children's Hospital, Florence
| | | | - Jennifer Heimall
- Perelman School of Medicine at University of Pennsylvania, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia
| | - Kathleen E Sullivan
- Perelman School of Medicine at University of Pennsylvania, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia
| | - Eric Allenspach
- Pediatric Immunology/Rheumatology, University of Washington, Seattle; Seattle Children's Hospital, Seattle
| | - Neil Romberg
- Perelman School of Medicine at University of Pennsylvania, Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia
| | - Sean G Deane
- Department of Allergy, The Permanente Medical Group, Sacramento, and the Division of Rheumatology/Allergy and Clinical Immunology, University of California, Davis, School of Medicine, Sacramento
| | - Benjamin T Prince
- Nationwide Children's Hospital Department of Allergy and Immunology, Columbus; College of Medicine, The Ohio State University, Columbus
| | - Melissa J Rose
- College of Medicine, The Ohio State University, Columbus; Division of Pediatric Hematology-Oncology, Nationwide Children's Hospital, Columbus
| | - John Bohnsack
- Department of Pediatrics, University of Utah, Salt Lake City
| | | | - Rohith Jesudas
- Department of Hematology, St Jude Children's Research Hospital, Memphis
| | - Maria Marluce Dos Santos Vilela
- Pediatric Allergy and Immunology/Center of Investigation in Pediatrics, Faculty of Medical Sciences, State University of Campinas-Unicamp, São Paulo
| | - Michael O'Sullivan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Immunology Department, Perth Children's Hospital, Nedlands
| | - Jana Pachlopnik Schmid
- Division of Immunology, University Children's Hospital Zurich, Children's Research Center (CRC), Zurich
| | - Štěpánka Průhová
- Department of Pediatrics, Charles University in Prague, Second Faculty of Medicine and University Hospital Motol, Prague
| | - Adam Klocperk
- Department of Immunology, Second Faculty of Medicine and University Hospital Motol, Charles University in Prague, Prague
| | - Matthew Rees
- Department of Hematology, St Jude Children's Research Hospital, Memphis
| | - Helen Su
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda
| | - Sami Bahna
- Allergy and Immunology Section, Louisiana State University Health Sciences Center, Shreveport
| | - Safa Baris
- School of Medicine, Pediatric Allergy and Immunology, Marmara University, Istanbul
| | - Lisa M Bartnikas
- Division of Immunology, Boston Children's Hospital, Department of Pediatrics, Harvard Medical School, Boston
| | - Amy Chang Berger
- Division of Hospital Medicine, Department of Medicine, University of California, San Francisco
| | - Tracy A Briggs
- Division of Evolution and Genomic Sciences, School of Biological Sciences, University of Manchester, Manchester; NW Genomic Laboratory Hub, Manchester Centre for Genomic Medicine, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester
| | - Shannon Brothers
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Starship Children's Hospital, Auckland
| | - Vanessa Bundy
- Allergy and Immunology, University of California, Los Angeles
| | - Alice Y Chan
- Department of Medicine, University of California, San Francisco
| | - Shanmuganathan Chandrakasan
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta
| | | | - Theresa Cole
- Department of Allergy and Immunology, The Royal Children's Hospital, Melbourne
| | - Matthew C Cook
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra
| | | | - Ute Fischer
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf
| | - David A Fulcher
- Department of Immunology and Infectious Diseases, John Curtin School of Medical Research, Australian National University, Canberra
| | - Silvanna Gallo
- Department of Pediatrics, Immunology and Rheumatology Section, Puerto Montt Hospital, Puerto Montt
| | - Amelie Gauthier
- Department of Allergy and Immunology, CHU de Québec-CHUL, Laval University Hospital Center, Laval University, Quebec City
| | - Andrew R Gennery
- Newcastle University Translational and Clinical Research Institute, Newcastle (United Kingdom)
| | - José Gonçalo Marques
- Infectious Diseases and Immunodeficiencies Unit, Department of Pediatrics, Hospital de Santa Maria-CHULN and Faculdade de Medicina, Universidade de Lisboa, Lisbon
| | - Frédéric Gottrand
- University Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, Lille
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Eyal Grunebaum
- Division of Immunology and Allergy, and the Department of Pediatrics, Developmental and Stem Cell Biology Program, Research Institute, Hospital for Sick Children, Toronto
| | - Emma Haapaniemi
- Centre for Molecular Medicine Norway, Oslo; Department of Pediatric Research, Oslo
| | | | - Kaarina Heiskanen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Turku and Kuopio, Finland
| | | | - Hal M Hoffman
- Department of Pediatrics, University of California San Diego, La Jolla; Rady Children's Hospital San Diego, Division of Pediatric Allergy, Immunology, and Rheumatology, San Diego
| | - Luis Ignacio Gonzalez-Granado
- Pediatrics Department, University Hospital 12 de Octubre, Research Institute Hospital, School of Medicine Complutense University, Madrid
| | - Anthony L Guerrerio
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore
| | - Leena Kainulainen
- Department of Pediatrics and Medicine, Turku University Hospital, University of Turku, Turku, Finland
| | - Ashish Kumar
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati
| | | | - Carina Levin
- Pediatric Hematology Unit, Emek Medical Centre, Afula, and the Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa
| | - Timi Martelius
- Adult Immunodeficiency Unit, Inflammation Center, Helsinki University Hospital and University of Helsinki, Helsinki
| | - Olaf Neth
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Peter Olbrich
- Pediatric Infectious Diseases, Rheumatology and Immunology Unit, Hospital Universitario Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain
| | - Alejandro Palma
- Servicio de Immunología y Reumatología, Hospital Nacional de Pediatría Prof Dr Juan P. Garrahan, Buenos Aires
| | - Niraj C Patel
- Division of Allergy and Immunology, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta
| | - Tamara Pozos
- Department of Clinical Immunology, Children's Minnesota, Minneapolis
| | - Kahn Preece
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Department of Paediatric Immunology, John Hunter Children's Hospital, Newcastle (Australia)
| | | | | | - Yael Schejter
- Department of Bone Marrow Transplantation and Cancer Immunotherapy, Hadassah Ein-Kerem Medical Center and Faculty of Medicine, Hebrew University, Jerusalem
| | - Christine Seroogy
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison
| | - Jan Sinclair
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Starship Children's Hospital, Auckland
| | - Effie Skevofilax
- Department of Pediatric Hematology-Oncology (TAO) and First Department of Pediatrics, Aghia Sophia Children's Hospital, Athens
| | - Daniel Suan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Garvan Institute of Medical Research, Darlinghurst; Westmead Clinical School, University of Sydney, Westmead
| | - Daniel Suez
- Allergy, Asthma & Immunology Clinic, PA, Irving
| | - Paul Szabolcs
- University of Pittsburgh Medical Center, Children's Hospital of Pittsburgh, Pittsburgh
| | - Helena Velasco
- Division of Allergy and Clinical Immunology, Moinhos de Vento Hospital, Porto Alegre
| | - Klaus Warnatz
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Kelly Walkovich
- Department of Pediatrics, C. S. Mott Children's Hospital, Michigan Medicine, Ann Arbor
| | - Austen Worth
- Great Ormond Street Hospital for Children, London
| | - Mikko R J Seppänen
- Rare Disease Center, Children's Hospital, and Adult Primary Immunodeficiency Outpatient Clinic, Inflammation Center, University of Helsinki and Helsinki University Hospital, Helsinki
| | | | - Georgios Sogkas
- Department of Clinical Immunology and Rheumatology, Hannover Medical School, Hanover
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg
| | - Stuart G Tangye
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA); Garvan Institute of Medical Research, Darlinghurst; St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney
| | - Megan A Cooper
- Department of Pediatrics, Division of Rheumatology and Immunology, Washington University School of Medicine, St Louis
| | - Joshua D Milner
- Department of Pediatrics, Division of Allergy and Immunology, Columbia University, New York Presbyterian Hospital, New York
| | - Lisa R Forbes Satter
- Department of Pediatrics, Baylor College of Medicine and William T. Shearer Center for Human Immunobiology, Texas Children's Hospital, Houston.
| |
Collapse
|
4
|
Wang W, Silva LM, Wang HH, Kavanaugh MA, Pottorf TS, Allard BA, Jacobs DT, Dong R, Cornelius JT, Chaturvedi A, Swenson-Fields KI, Fields TA, Pritchard MT, Sharma M, Slawson C, Wallace DP, Calvet JP, Tran PV. Ttc21b deficiency attenuates autosomal dominant polycystic kidney disease in a kidney tubular- and maturation-dependent manner. Kidney Int 2022; 102:577-591. [PMID: 35644283 PMCID: PMC9398994 DOI: 10.1016/j.kint.2022.04.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 04/21/2022] [Accepted: 04/29/2022] [Indexed: 01/26/2023]
Abstract
Primary cilia are sensory organelles built and maintained by intraflagellar transport (IFT) multiprotein complexes. Deletion of several IFT-B genes attenuates polycystic kidney disease (PKD) severity in juvenile and adult autosomal dominant polycystic kidney disease (ADPKD) mouse models. However, deletion of an IFT-A adaptor, Tulp3, attenuates PKD severity in adult mice only. These studies indicate that dysfunction of specific cilia components has potential therapeutic value. To broaden our understanding of cilia dysfunction and its therapeutic potential, we investigate the role of global deletion of an IFT-A gene, Ttc21b, in juvenile and adult mouse models of ADPKD. Both juvenile (postnatal day 21) and adult (six months of age) ADPKD mice exhibited kidney cysts, increased kidney weight/body weight ratios, lengthened kidney cilia, inflammation, and increased levels of the nutrient sensor, O-linked β-N-acetylglucosamine (O-GlcNAc). Deletion of Ttc21b in juvenile ADPKD mice reduced cortical collecting duct cystogenesis and kidney weight/body weight ratios, increased proximal tubular and glomerular dilations, but did not reduce cilia length, inflammation, nor O-GlcNAc levels. In contrast, Ttc21b deletion in adult ADPKD mice markedly attenuated kidney cystogenesis and reduced cilia length, inflammation, and O-GlcNAc levels. Thus, unlike IFT-B, the effect of Ttc21b deletion in mouse models of ADPKD is development-specific. Unlike an IFT-A adaptor, deleting Ttc21b in juvenile ADPKD mice is partially ameliorative. Thus, our studies suggest that different microenvironmental factors, found in distinct nephron segments and in developing versus mature stages, modify ciliary homeostasis and ADPKD pathobiology. Further, elevated levels of O-GlcNAc, which regulates cellular metabolism and ciliogenesis, may be a pathological feature of ADPKD.
Collapse
Affiliation(s)
- Wei Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Luciane M Silva
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Henry H Wang
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Matthew A Kavanaugh
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Tana S Pottorf
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Bailey A Allard
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Damon T Jacobs
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Rouchen Dong
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Joseph T Cornelius
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Aakriti Chaturvedi
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Katherine I Swenson-Fields
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Timothy A Fields
- Department of Pathology and Laboratory Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Michele T Pritchard
- Pharmacology, Toxicology and Therapeutics, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Madhulika Sharma
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Chad Slawson
- Department of Biochemistry and Molecular Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Darren P Wallace
- Department of Internal Medicine, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - James P Calvet
- Department of Biochemistry and Molecular Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Pamela V Tran
- Department of Anatomy and Cell Biology, The Jared Grantham Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas, USA.
| |
Collapse
|
5
|
Park JY, Yoo KD, Bae E, Kim KH, Lee JW, Shin SJ, Lee JS, Kim YS, Yang SH. Blockade of STAT3 signaling alleviates progression of acute kidney injury-to-chronic kidney disease through anti-apoptosis. Am J Physiol Renal Physiol 2022; 322:F553-F572. [PMID: 35311382 DOI: 10.1152/ajprenal.00595.2020] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a pivotal mediator of IL-6-type cytokine signaling. However, the roles of its full-length and truncated isoforms in acute kidney injury (AKI) and its transition to chronic kidney disease (CKD) remain elusive. Herein, the role of STAT3 isoforms in AKI-to-CKD transition was characterized using an ischemia-reperfusion injury (IRI) mouse model. IRI was induced in C57BL/6 mice. Stattic®, a STAT3 inhibitor, was administered to the mice 3 h prior to IRI. Intrarenal cytokine expression was quantified using real-time PCR, and FACS analysis was performed. The effect of Stattic® on human tubular epithelial cells (TECs) cultured under hypoxic conditions was also evaluated. Phosphorylated STAT3 isoforms were detected by western blotting. Stattic® treatment attenuated IRI-induced tubular damage and inflammatory cytokine/chemokine expression, while decreasing macrophage infiltration and fibrosis in mouse unilateral IRI and UUO models. Similarly, in vitro STAT3 inhibition downregulated fibrosis and apoptosis in 72-h hypoxia-induced human TECs and reduced pSTAT3α-mediated inflammation. Moreover, pSTAT3 expression was increased in human acute tubular necrosis and CKD tissues. STAT3 activation is associated with IRI progression, and STAT3-α may be a significant contributor. Hence, STAT3 may affect AKI-to-CKD transition, suggesting a novel strategy for AKI management with STAT3 inhibitors.
Collapse
Affiliation(s)
- Jae Yoon Park
- Department of Internal Medicine, Dongguk University College of Medicine, Dongguk University Ilsan Hospital, Goyang-si, Korea (South), Republic of
| | - Kyung Don Yoo
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea (South), Republic of
| | - Eunjin Bae
- Department of Internal Medicine, Gyeongsang National University College of Medicine, Gyeongsang University Changwon Hospital, Changwon, Korea (South), Republic of
| | - Kyu Hong Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea (South), Republic of
| | - Jae Wook Lee
- Nephrology Clinic, National Cancer Center of Korea, Seoul, Korea (South), Republic of
| | - Sung Joon Shin
- Department of Internal Medicine, Dongguk University College of Medicine, Dongguk University Ilsan Hospital, Goyang-si, Korea (South), Republic of
| | - Jong Soo Lee
- Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea (South), Republic of
| | - Yon Su Kim
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea (South), Republic of.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea (South), Republic of.,Kidney Research Institute, Seoul National University, Seoul, Korea (South), Republic of.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea (South), Republic of
| | - Seung Hee Yang
- Kidney Research Institute, Seoul National University, Seoul, Korea (South), Republic of.,Biomedical Research Institute, Seoul National University Hospital, Seoul, Korea (South), Republic of
| |
Collapse
|
6
|
Ppia is the most stable housekeeping gene for qRT-PCR normalization in kidneys of three Pkd1-deficient mouse models. Sci Rep 2021; 11:19798. [PMID: 34611276 PMCID: PMC8492864 DOI: 10.1038/s41598-021-99366-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/13/2021] [Indexed: 01/08/2023] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) is the most common inherited renal disorder, characterized by renal cyst development leading to end-stage renal disease. Although the appropriate choice of suitable reference is critical for quantitative RNA analysis, no comparison of frequently used “housekeeping” genes is available. Here, we determined the validity of 7 candidate housekeeping genes (Actb, Actg1, B2m, Gapdh, Hprt, Pgam1 and Ppia) in kidney tissues from mouse models orthologous to ADPKD, including a cystic mice (CY) 10–12 weeks old (Pkd1flox/flox:Nestincre/Pkd1flox/−:Nestincre, n = 10) and non-cystic (NC) controls (Pkd1flox/flox/Pkd1flox/-, n = 10), Pkd1-haploinsufficient (HT) mice (Pkd1+/−, n = 6) and wild-type (WT) controls (Pkd1+/+, n = 6) and a severely cystic (SC) mice 15 days old (Pkd1V/V, n = 7) and their controls (CO, n = 5). Gene expression data were analyzed using six distinct statistical softwares. The estimation of the ideal number of genes suggested the use of Ppia alone as sufficient, although not ideal, to analyze groups altogether. Actb, Hprt and Ppia expression profiles were correlated in all samples. Ppia was identified as the most stable housekeeping gene, while Gapdh was the least stable for all kidney samples. Stat3 expression level was consistent with upregulation in SC compared to CO when normalized by Ppia expression. In conclusion, present findings identified Ppia as the best housekeeping gene for CY + NC and SC + CO groups, while Hprt was the best for the HT + WT group.
Collapse
|
7
|
Pyrimethamine conjugated histone deacetylase inhibitors: Design, synthesis and evidence for triple negative breast cancer selective cytotoxicity. Bioorg Med Chem 2020; 28:115345. [DOI: 10.1016/j.bmc.2020.115345] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 01/12/2020] [Accepted: 01/22/2020] [Indexed: 12/25/2022]
|
8
|
Winokurow N, Schumacher S. A role for polycystin-1 and polycystin-2 in neural progenitor cell differentiation. Cell Mol Life Sci 2019; 76:2851-2869. [PMID: 30895336 PMCID: PMC11105687 DOI: 10.1007/s00018-019-03072-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/17/2019] [Accepted: 03/14/2019] [Indexed: 10/27/2022]
Abstract
Polycystin-1 (PC1) and polycystin-2 (PC2) are transmembrane proteins encoded by the Pkd1 and Pkd2 genes, respectively. Mutations in these genes are causative for the development of autosomal-dominant polycystic kidney disease. A prominent feature of this disease is an unbalanced cell proliferation. PC1 and PC2 physically interact to form a complex, which localizes to the primary cilia of renal epithelial cells. Recently, PC1 and PC2 have also been described to be present in primary cilia of radial glial cells (RGCs) and to contribute to the planar cell polarity of late RGCs and E1 ependymal cells. As neural progenitor cells (NPCs), early RGCs have to balance proliferation for expansion, or for self-renewal and differentiation to generate neurons. It is not known whether the polycystins play a role in this process. Here, we show that PC1 and PC2 are expressed in RGCs of the developing mouse cerebral cortex during neurogenesis. Loss-of-function analysis and cell-based assays reveal that a reduction of PC1 or PC2 expression leads to increased NPC proliferation, while the differentiation to neurons becomes impaired. The increased NPC proliferation is preceded by enhanced Notch signaling and accompanied by a rise in the number of symmetric cell divisions. The transcription factor STAT3 seems to be mechanistically important for polycystin signaling in NPCs as either STAT3 knockdown or inhibition of STAT3 function abrogates the increased proliferation driven by reduced polycystin expression. Our findings indicate that PC1 and PC2 are critical for maintaining a balance between proliferation and differentiation of NPCs.
Collapse
Affiliation(s)
- Natalie Winokurow
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany
| | - Stefan Schumacher
- Institute of Molecular and Cellular Anatomy, Ulm University, Albert-Einstein-Allee 11, 89081, Ulm, Germany.
| |
Collapse
|
9
|
Papavassiliou KA, Zoi I, Gargalionis AN, Koutsilieris M. Polycystin-1 affects cancer cell behaviour and interacts with mTOR and Jak signalling pathways in cancer cell lines. J Cell Mol Med 2019; 23:6215-6227. [PMID: 31251475 PMCID: PMC6714176 DOI: 10.1111/jcmm.14506] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 01/28/2023] Open
Abstract
Polycystic Kidney Disease (PKD), which is attributable to mutations in the PKD1 and PKD2 genes encoding polycystin‐1 (PC1) and polycystin‐2 (PC2) respectively, shares common cellular defects with cancer, such as uncontrolled cell proliferation, abnormal differentiation and increased apoptosis. Interestingly, PC1 regulates many signalling pathways including Jak/STAT, mTOR, Wnt, AP‐1 and calcineurin‐NFAT which are also used by cancer cells for sending signals that will allow them to acquire and maintain malignant phenotypes. Nevertheless, the molecular relationship between polycystins and cancer is unknown. In this study, we investigated the role of PC1 in cancer biology using glioblastoma (GOS3), prostate (PC3), breast (MCF7), lung (A549) and colorectal (HT29) cancer cell lines. Our in vitro results propose that PC1 promotes cell migration in GOS3 cells and suppresses cell migration in A549 cells. In addition, PC1 enhances cell proliferation in GOS3 cells but inhibits it in MCF7, A549 and HT29 cells. We also found that PC1 up‐regulates mTOR signalling and down‐regulates Jak signalling in GOS3 cells, while it up‐regulates mTOR signalling in PC3 and HT29 cells. Together, our study suggests that PC1 modulates cell proliferation and migration and interacts with mTOR and Jak signalling pathways in different cancer cell lines. Understanding the molecular details of how polycystins are associated with cancer may lead to the identification of new players in this devastating disease.
Collapse
Affiliation(s)
- Kostas A Papavassiliou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ilianna Zoi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios N Gargalionis
- Department of Biopathology, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
10
|
Formica C, Happé H, Veraar KA, Vortkamp A, Scharpfenecker M, McNeill H, Peters DJ. Four-jointed knock-out delays renal failure in an ADPKD model with kidney injury. J Pathol 2019; 249:114-125. [PMID: 31038742 PMCID: PMC6772084 DOI: 10.1002/path.5286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/19/2019] [Accepted: 04/26/2019] [Indexed: 12/28/2022]
Abstract
Autosomal Dominant Polycystic Kidney Disease is characterised by the development of fluid‐filled cysts in the kidneys which lead to end‐stage renal disease (ESRD). In the majority of cases, the disease is caused by a mutation in the Pkd1 gene. In a previous study, we demonstrated that renal injury can accelerate cyst formation in Pkd1 knock‐out (KO) mice. In that study, we found that after injury four‐jointed (Fjx1), an upstream regulator of planar cell polarity and the Hippo pathway, was aberrantly expressed in Pkd1 KO mice compared to WT. Therefore, we hypothesised a role for Fjx1 in injury/repair and cyst formation. We generated single and double deletion mice for Pkd1 and Fjx1, and we induced toxic renal injury using the nephrotoxic compound 1,2‐dichlorovinyl‐cysteine. We confirmed that nephrotoxic injury can accelerate cyst formation in Pkd1 mutant mice. This caused Pkd1 KO mice to reach ESRD significantly faster; unexpectedly, double KO mice survived significantly longer. Cyst formation was comparable in both models, but we found significantly less fibrosis and macrophage infiltration in double KO mice. Taken together, these data suggest that Fjx1 disruption protects the cystic kidneys against kidney failure by reducing inflammation and fibrosis. Moreover, we describe, for the first time, an interesting (yet unidentified) mechanism that partially discriminates cyst growth from fibrogenesis. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Chiara Formica
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Hester Happé
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Kimberley Am Veraar
- Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrea Vortkamp
- Department of Developmental Biology, Centre of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | | | - Helen McNeill
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | - Dorien Jm Peters
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
11
|
Patera F, Cudzich-Madry A, Huang Z, Fragiadaki M. Renal expression of JAK2 is high in polycystic kidney disease and its inhibition reduces cystogenesis. Sci Rep 2019; 9:4491. [PMID: 30872773 PMCID: PMC6418191 DOI: 10.1038/s41598-019-41106-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 02/28/2019] [Indexed: 12/21/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is the most common renal genetic disorder, however it still lacks a cure. The discovery of new therapies heavily depends on understanding key signalling pathways that lead to ADPKD. The JAnus Kinase and Signal Transducers and Activators of Transcription (JAK/STAT) pathway is aberrantly activated and contributes to ADPKD pathogenesis via enhancing epithelial proliferation. Yet the mechanisms underlying the upregulation of JAK/STAT activity in this disease context is completely unknown. Here, we investigate the role of JAK2 in ADPKD using a murine model of ADPKD (Pkd1nl/nl). In normal kidneys, JAK2 expression is limited to tubular epithelial and vascular cells with lesser staining in bowman’s capsule and remains below detection level in the interstitium. By contrast, in kidneys of mice with ADPKD, JAK2 is higher in cyst-lining cells when compared to normal tubules and critically, it is ectopically expressed in the interstitium, suggesting that ectopic JAK2 may contribute to ADPKD. JAK2 activity was inhibited using either curcumin, a natural compound with strong JAK2 inhibitor activity, or Tofacitinib, a clinically used selective JAK small molecule inhibitor. JAK2 inhibition led to significantly reduced tyrosine phosphorylation of STAT3 and markedly reduced cystic growth of human and mouse ADPKD-derived cells in cystogenesis assays. Taken together, our results indicate that blockade of JAK2 shows promise as a novel therapeutic target in ADPKD.
Collapse
Affiliation(s)
- Foteini Patera
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, United Kingdom
| | - Alex Cudzich-Madry
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, United Kingdom
| | - Zhi Huang
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, United Kingdom
| | - Maria Fragiadaki
- Academic Nephrology Unit, Department of Infection, Immunity and Cardiovascular Disease, University of Sheffield, Sheffield, S10 2RX, United Kingdom.
| |
Collapse
|
12
|
Robinson RL, Sharma A, Bai S, Heneidi S, Lee TJ, Kodeboyina SK, Patel N, Sharma S. Comparative STAT3-Regulated Gene Expression Profile in Renal Cell Carcinoma Subtypes. Front Oncol 2019; 9:72. [PMID: 30863721 PMCID: PMC6399114 DOI: 10.3389/fonc.2019.00072] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/25/2019] [Indexed: 12/15/2022] Open
Abstract
Renal cell carcinomas (RCC) are heterogeneous and can be further classified into three major subtypes including clear cell, papillary and chromophobe. Signal transducer and activator of transcription 3 (STAT3) is commonly hyperactive in many cancers and is associated with cancer cell proliferation, invasion, migration, and angiogenesis. In renal cell carcinoma, increased STAT3 activation is associated with increased metastasis and worse survival outcomes, but clinical trials targeting the STAT3 signaling pathway have shown varying levels of success in different RCC subtypes. Using RNA-seq data from The Cancer Genome Atlas (TCGA), we compared expression of 32 STAT3 regulated genes in 3 RCC subtypes. Our results indicate that STAT3 activation plays the most significant role in clear cell RCC relative to the other subtypes, as half of the evaluated genes were upregulated in this subtype. MMP9, BIRC5, and BCL2 were upregulated and FOS was downregulated in all three subtypes. Several genes including VEGFA, VIM, MYC, ITGB4, ICAM1, MMP1, CCND1, STMN1, TWIST1, and PIM2 had variable expression in RCC subtypes and are potential therapeutic targets for personalized medicine.
Collapse
Affiliation(s)
- Rebekah L Robinson
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ashok Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Population Health Sciences, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Shan Bai
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Saleh Heneidi
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Tae Jin Lee
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sai Karthik Kodeboyina
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Nikhil Patel
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Shruti Sharma
- Center for Biotechnology and Genomic Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
13
|
Weimbs T, Shillingford JM, Torres J, Kruger SL, Bourgeois BC. Emerging targeted strategies for the treatment of autosomal dominant polycystic kidney disease. Clin Kidney J 2018; 11:i27-i38. [PMID: 30581563 PMCID: PMC6295603 DOI: 10.1093/ckj/sfy089] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 08/27/2018] [Indexed: 12/25/2022] Open
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a widespread genetic disease that leads to renal failure in the majority of patients. The very first pharmacological treatment, tolvaptan, received Food and Drug Administration approval in 2018 after previous approval in Europe and other countries. However, tolvaptan is moderately effective and may negatively impact a patient's quality of life due to potentially significant side effects. Additional and improved therapies are still urgently needed, and several clinical trials are underway, which are discussed in the companion paper Müller and Benzing (Management of autosomal-dominant polycystic kidney disease-state-of-the-art) Clin Kidney J 2018; 11: i2-i13. Here, we discuss new therapeutic avenues that are currently being investigated at the preclinical stage. We focus on mammalian target of rapamycin and dual kinase inhibitors, compounds that target inflammation and histone deacetylases, RNA-targeted therapeutic strategies, glucosylceramide synthase inhibitors, compounds that affect the metabolism of renal cysts and dietary restriction. We discuss tissue targeting to renal cysts of small molecules via the folate receptor, and of monoclonal antibodies via the polymeric immunoglobulin receptor. A general problem with potential pharmacological approaches is that the many molecular targets that have been implicated in ADPKD are all widely expressed and carry out important functions in many organs and tissues. Because ADPKD is a slowly progressing, chronic disease, it is likely that any therapy will have to continue over years and decades. Therefore, systemically distributed drugs are likely to lead to potentially prohibitive extra-renal side effects during extended treatment. Tissue targeting to renal cysts of such drugs is one potential way around this problem. The use of dietary, instead of pharmacological, interventions is another.
Collapse
Affiliation(s)
- Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Jonathan M Shillingford
- Department of Internal Medicine, Division of Nephrology, University of Michigan, Ann Arbor, MI, USA
| | - Jacob Torres
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Samantha L Kruger
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| | - Bryan C Bourgeois
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute, University of California, Santa Barbara, CA, USA
| |
Collapse
|
14
|
Renal Injury during Long-Term Crizotinib Therapy. Int J Mol Sci 2018; 19:ijms19102902. [PMID: 30257437 PMCID: PMC6213486 DOI: 10.3390/ijms19102902] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/03/2018] [Accepted: 09/11/2018] [Indexed: 01/01/2023] Open
Abstract
Crizotinib is highly effective against anaplastic lymphoma kinase-positive and c-ros oncogen1-positive non-small cell lung cancer. Renal dysfunction is associated with crizotinib therapy but the mechanism is unknown. Here, we report a case of anaplastic lymphoma kinase positive non-small cell lung cancer showing multiple cysts and dysfunction of the kidneys during crizotinib administration. We also present results demonstrating that long-term crizotinib treatment induces fibrosis and dysfunction of the kidneys by activating the tumor necrosis factor-α/nuclear factor-κB signaling pathway. In conclusion, this study shows the renal detrimental effects of crizotinib, suggesting the need of careful monitoring of renal function during crizotinib therapy.
Collapse
|
15
|
Aghakhani Chegeni S, Rahimzadeh M, Montazerghaem H, Khayatian M, Dasturian F, Naderi N. Preliminary Report on the Association Between STAT3 Polymorphisms and Susceptibility to Acute Kidney Injury After Cardiopulmonary Bypass. Biochem Genet 2018; 56:627-638. [PMID: 29846833 DOI: 10.1007/s10528-018-9865-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 05/18/2018] [Indexed: 01/01/2023]
Abstract
Cardiopulmonary bypass-associated acute kidney injury (CPB-AKI) is a well-recognized complication which is clearly linked to increased morbidity and mortality. Due to important role of inflammation in CPB-AKI pathogenesis, we explored the association between polymorphisms in STAT3, an inflammation-associated transcription factor, and the risk of CPB-AKI. In this study, STAT3 rs1053004 and rs744166 polymorphisms were analyzed in 129 patients undergoing coronary artery bypass grafting in Jorjani heart center, Bandar Abbas, Iran. The genotypes were determined using sequence-specific primers (PCR-SSP). Sixty-three patients met the criteria for AKI after cardiac surgery (AKI group). The remaining 66 patients did not develop AKI (non-AKI group). Rs1053004 GG genotype was significantly associated with a decreased risk (OR 0.4, 95% CI 0.17-0.9, P = 0.03) of CPB-AKI. Subgroup analyses revealed that GG genotype has also a protective effect in older patients (Age ≥ 60) (OR 0.19, 95% CI 0.04-0.8, P = 0.01). However, rs744166 did not show any difference between AKI and non-AKI groups. The result of our study for the first time provides evidence that rs1053004 polymorphism is significantly associated with a decreased risk of CPB-AKI in Iranian population, especially in older subjects.
Collapse
Affiliation(s)
- Sara Aghakhani Chegeni
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.,Food Health Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahsa Rahimzadeh
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Hossein Montazerghaem
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mahmood Khayatian
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Farzaneh Dasturian
- Department of Biochemistry, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nadereh Naderi
- Cardiovascular Research Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran. .,Department of Immunology, Faculty of Medicine, Hormozgan University of Medical Sciences, EmamHossein Boulevard, P.O. Box: 7919693116, Bandar Abbas, Iran.
| |
Collapse
|
16
|
Li J, Lu D, Liu H, Williams BO, Overbeek PA, Lee B, Zheng L, Yang T. Sclt1 deficiency causes cystic kidney by activating ERK and STAT3 signaling. Hum Mol Genet 2018; 26:2949-2960. [PMID: 28486600 DOI: 10.1093/hmg/ddx183] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 05/03/2017] [Indexed: 12/31/2022] Open
Abstract
Ciliopathies form a group of inherited disorders sharing several clinical manifestations because of abnormal cilia formation or function, and few treatments have been successful against these disorders. Here, we report a mouse model with mutated Sclt1 gene, which encodes a centriole distal appendage protein important for ciliogenesis. Sodium channel and clathrin linker 1 (SCLT1) mutations were associated with the oral-facial-digital syndrome (OFD), an autosomal recessive ciliopathy. The Sclt1-/- mice exhibit typical ciliopathy phenotypes, including cystic kidney, cleft palate and polydactyly. Sclt1-loss decreases the number of cilia in kidney; increases proliferation and apoptosis of renal tubule epithelial cells; elevates protein kinase A, extracellular signal-regulated kinases, SMAD and signal transducer and activator of transcription 3 (STAT3) pathways; and enhances pro-inflammation and pro-fibrosis pathways with disease progression. Embryonic kidney cyst formation of Sclt1-/- mice was effectively reduced by an anti-STAT3 treatment using pyrimethamine. Overall, we reported a new mouse model for the OFD; and our data suggest that STAT3 inhibition may be a promising treatment for SCLT1-associated cystic kidney.
Collapse
Affiliation(s)
- Jianshuang Li
- Hubei Key Laboratory of Cell Homeostasis, Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China.,Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Di Lu
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Huadie Liu
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Bart O Williams
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | | | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, Department of Cell Biology, College of Life Sciences, Wuhan University, Wuhan, Hubei 430072, P.R. China
| | - Tao Yang
- Program for Skeletal Disease and Tumor Metastasis, Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|
17
|
Yokota T, Omachi K, Suico MA, Kamura M, Kojima H, Fukuda R, Motomura K, Teramoto K, Kaseda S, Kuwazuru J, Takeo T, Nakagata N, Shuto T, Kai H. STAT3 inhibition attenuates the progressive phenotypes of Alport syndrome mouse model. Nephrol Dial Transplant 2017; 33:214-223. [DOI: 10.1093/ndt/gfx246] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 06/26/2017] [Indexed: 11/12/2022] Open
Affiliation(s)
- Tsubasa Yokota
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Kohei Omachi
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
- Program for Leading Graduate School Health Life Science: Interdisciplinary and Glocal Oriented Program, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Mary Ann Suico
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Misato Kamura
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
- Program for Leading Graduate School Health Life Science: Interdisciplinary and Glocal Oriented Program, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Haruka Kojima
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Ryosuke Fukuda
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Keishi Motomura
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Keisuke Teramoto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
- Program for Leading Graduate School Health Life Science: Interdisciplinary and Glocal Oriented Program, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Shota Kaseda
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
- Program for Leading Graduate School Health Life Science: Interdisciplinary and Glocal Oriented Program, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Jun Kuwazuru
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto City, Kumamoto, Japan
- Program for Leading Graduate School Health Life Science: Interdisciplinary and Glocal Oriented Program, Kumamoto University, Kumamoto City, Kumamoto, Japan
| |
Collapse
|
18
|
Song CJ, Zimmerman KA, Henke SJ, Yoder BK. Inflammation and Fibrosis in Polycystic Kidney Disease. Results Probl Cell Differ 2017; 60:323-344. [PMID: 28409351 PMCID: PMC7875307 DOI: 10.1007/978-3-319-51436-9_12] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Polycystic kidney disease (PKD) is a commonly inherited disorder characterized by cyst formation and fibrosis (Wilson, N Engl J Med 350:151-164, 2004) and is caused by mutations in cilia or cilia-related proteins, such as polycystin 1 or 2 (Oh and Katsanis, Development 139:443-448, 2012; Kotsis et al., Nephrol Dial Transplant 28:518-526, 2013). A major pathological feature of PKD is the development of interstitial inflammation and fibrosis with an associated accumulation of inflammatory cells (Grantham, N Engl J Med 359:1477-1485, 2008; Zeier et al., Kidney Int 42:1259-1265, 1992; Ibrahim, Sci World J 7:1757-1767, 2007). It is unclear whether inflammation is a driving force for cyst formation or a consequence of the pathology (Ta et al., Nephrology 18:317-330, 2013) as in some murine models cysts are present prior to the increase in inflammatory cells (Phillips et al., Kidney Blood Press Res 30:129-144, 2007; Takahashi et al., J Am Soc Nephrol JASN 1:980-989, 1991), while in other models the increase in inflammatory cells is present prior to or coincident with cyst initiation (Cowley et al., Kidney Int 43:522-534, 1993, Kidney Int 60:2087-2096, 2001). Additional support for inflammation as an important contributor to cystic kidney disease is the increased expression of many pro-inflammatory cytokines in murine models and human patients with cystic kidney disease (Karihaloo et al., J Am Soc Nephrol JASN 22:1809-1814, 2011; Swenson-Fields et al., Kidney Int, 2013; Li et al., Nat Med 14:863-868, 2008a). Based on these data, an emerging model in the field is that disruption of primary cilia on tubule epithelial cells leads to abnormal cytokine cross talk between the epithelium and the inflammatory cells contributing to cyst growth and fibrosis (Ta et al., Nephrology 18:317-330, 2013). These cytokines are produced by interstitial fibroblasts, inflammatory cells, and tubule epithelial cells and activate multiple pathways including the JAK-STAT and NF-κB signaling (Qin et al., J Am Soc Nephrol JASN 23:1309-1318, 2012; Park et al., Am J Nephrol 32:169-178, 2010; Bhunia et al., Cell 109:157-168, 2002). Indeed, inflammatory cells are responsible for producing several of the pro-fibrotic growth factors observed in PKD patients with fibrosis (Nakamura et al., Am J Nephrol 20:32-36, 2000; Wilson et al., J Cell Physiol 150:360-369, 1992; Song et al., Hum Mol Genet 18:2328-2343, 2009; Schieren et al., Nephrol Dial Transplant 21:1816-1824, 2006). These growth factors trigger epithelial cell proliferation and myofibroblast activation that stimulate the production of extracellular matrix (ECM) genes including collagen types 1 and 3 and fibronectin, leading to reduced glomerular function with approximately 50% of ADPKD patients progressing to end-stage renal disease (ESRD). Therefore, treatments designed to reduce inflammation and slow the rate of fibrosis are becoming important targets that hold promise to improve patient life span and quality of life. In fact, recent studies in several PKD mouse models indicate that depletion of macrophages reduces cyst severity. In this chapter, we review the potential mechanisms of interstitial inflammation in PKD with a focus on ADPKD and discuss the role of interstitial inflammation in progression to fibrosis and ESRD.
Collapse
Affiliation(s)
- Cheng Jack Song
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kurt A Zimmerman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Scott J Henke
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Bradley K Yoder
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
19
|
STAT5 drives abnormal proliferation in autosomal dominant polycystic kidney disease. Kidney Int 2017; 91:575-586. [PMID: 28104302 DOI: 10.1016/j.kint.2016.10.039] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 10/10/2016] [Accepted: 10/27/2016] [Indexed: 11/23/2022]
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) leads to renal failure. The hallmark of ADPKD is increased epithelial proliferation, which has been proposed to be due to atypical signaling including abnormal JAK-STAT activity. However, the relative contribution of JAK-STAT family members in promoting proliferation in ADPKD is unknown. Here, we present siRNA JAK-STAT-focused screens discovering a previously unknown proliferative role for multiple JAK-STAT components (including STAT1, STAT2, STAT4, STAT5a, and STAT5b). Amongst these, we selected to study the growth hormone/growth hormone receptor/STAT5-axis because of its known role as a regulator of growth in nonrenal tissues. Loss of STAT5 function, facilitated by pharmacological inhibition or siRNAs, significantly reduced proliferation with an associated reduction in cyst growth in vitro. To study whether STAT5 is abnormally activated in vivo, we analyzed its expression using two independent mouse models of ADPKD. STAT5 was nuclear, thus activated, in renal epithelial cyst lining cells in both models. To test whether forced activation of STAT5 can modulate proliferation of renal cells in vivo, irrespective of the Pkd1 status, we overexpressed growth hormone. These mice showed increased STAT5 activity in renal epithelial cells, which correlated with de novo expression of cyclin D1, a STAT5 target gene. Chromatin immunoprecipitation experiments revealed that STAT5 transcriptionally activated cyclin D1 in a growth hormone-dependent fashion, thus providing a mechanism into how STAT5 enhances proliferation. Finally, we provide evidence of elevated serum growth hormone in Pkd1 mutant mice. Thus, the growth hormone/STAT5 signaling axis is a novel therapeutic target in ADPKD.
Collapse
|
20
|
Abstract
Autosomal dominant polycystic kidney disease (ADPKD) is a signalopathy of renal tubular epithelial cells caused by naturally occurring mutations in two distinct genes, polycystic kidney disease 1 (PKD1) and 2 (PKD2). Genetic variants in PKD1, which encodes the polycystin-1 (PC-1) protein, remain the predominant factor associated with the pathogenesis of nearly two-thirds of all patients diagnosed with PKD. Although the relationship between defective PC-1 with renal cystic disease initiation and progression remains to be fully elucidated, there are numerous clinical studies that have focused upon the control of effector systems involving heterotrimeric G protein regulation. A major regulator in the activation state of heterotrimeric G proteins are G protein-coupled receptors (GPCRs), which are defined by their seven transmembrane-spanning regions. PC-1 has been considered to function as an unconventional GPCR, but the mechanisms by which PC-1 controls signal processing, magnitude, or trafficking through heterotrimeric G proteins remains to be fully known. The diversity of heterotrimeric G protein signaling in PKD is further complicated by the presence of non-GPCR proteins in the membrane or cytoplasm that also modulate the functional state of heterotrimeric G proteins within the cell. Moreover, PC-1 abnormalities promote changes in hormonal systems that ultimately interact with distinct GPCRs in the kidney to potentially amplify or antagonize signaling output from PC-1. This review will focus upon the canonical and noncanonical signaling pathways that have been described in PKD with specific emphasis on which heterotrimeric G proteins are involved in the pathological reorganization of the tubular epithelial cell architecture to exacerbate renal cystogenic pathways.
Collapse
Affiliation(s)
- Taketsugu Hama
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| | - Frank Park
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee
| |
Collapse
|
21
|
Mormile R, Fanos V, Vittori G. Preterm infants, kidney, rickets and vitamin D intake: is it time for rewriting the history? Arch Gynecol Obstet 2014; 290:1055-1057. [PMID: 25151029 DOI: 10.1007/s00404-014-3425-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 08/11/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Raffaella Mormile
- Division of Pediatrics and Neonatology, Moscati Hospital, Via A. Gramsci, 3, 81031, Aversa, Italy,
| | | | | |
Collapse
|
22
|
Talbot JJ, Song X, Wang X, Rinschen MM, Doerr N, LaRiviere WB, Schermer B, Pei YP, Torres VE, Weimbs T. The cleaved cytoplasmic tail of polycystin-1 regulates Src-dependent STAT3 activation. J Am Soc Nephrol 2014; 25:1737-48. [PMID: 24578126 PMCID: PMC4116067 DOI: 10.1681/asn.2013091026] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 12/06/2013] [Indexed: 12/30/2022] Open
Abstract
Polycystin-1 (PC1) mutations result in proliferative renal cyst growth and progression to renal failure in autosomal dominant polycystic kidney disease (ADPKD). The transcription factor STAT3 (signal transducer and activator of transcription 3) was shown to be activated in cyst-lining cells in ADPKD and PKD mouse models and may drive renal cyst growth, but the mechanisms leading to persistent STAT3 activation are unknown. A proteolytic fragment of PC1 corresponding to the cytoplasmic tail, PC1-p30, is overexpressed in ADPKD. Here, we show that PC1-p30 interacts with the nonreceptor tyrosine kinase Src, resulting in Src-dependent activation of STAT3 by tyrosine phosphorylation. The PC1-p30-mediated activation of Src/STAT3 was independent of JAK family kinases and insensitive to the STAT3 inhibitor suppressor of cytokine signaling 3. Signaling by the EGF receptor (EGFR) or cAMP amplified the activation of Src/STAT3 by PC1-p30. Expression of PC1-p30 changed the cellular response to cAMP signaling. In the absence of PC1-p30, cAMP dampened EGFR- or IL-6-dependent activation of STAT3; in the presence of PC1-p30, cAMP amplified Src-dependent activation of STAT3. In the polycystic kidney (PCK) rat model, activation of STAT3 in renal cystic cells depended on vasopressin receptor 2 (V2R) signaling, which increased cAMP levels. Genetic inhibition of vasopressin expression or treatment with a pharmacologic V2R inhibitor strongly suppressed STAT3 activation and reduced renal cyst growth. These results suggest that PC1, via its cleaved cytoplasmic tail, integrates signaling inputs from EGFR and cAMP, resulting in Src-dependent activation of STAT3 and a proliferative response.
Collapse
Affiliation(s)
- Jeffrey J Talbot
- Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California
| | - Xuewen Song
- Divisions of Nephrology and Genomic Medicine, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Xiaofang Wang
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Markus M Rinschen
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Cologne, Germany
| | - Nicholas Doerr
- Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California
| | - Wells B LaRiviere
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, Cologne, Germany; Systems Biology of Aging Cologne (Sybacol), Cologne, Germany; and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - York P Pei
- Divisions of Nephrology and Genomic Medicine, University Health Network and University of Toronto, Toronto, Ontario, Canada
| | - Vicente E Torres
- Division of Nephrology and Hypertension, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology, and Neuroscience Research Institute, University of California Santa Barbara, Santa Barbara, California;
| |
Collapse
|
23
|
Weimbs T, Olsan EE, Talbot JJ. Regulation of STATs by polycystin-1 and their role in polycystic kidney disease. JAKSTAT 2014; 2:e23650. [PMID: 24058808 PMCID: PMC3710321 DOI: 10.4161/jkst.23650] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Revised: 01/16/2013] [Accepted: 01/17/2013] [Indexed: 01/14/2023] Open
Abstract
Autosomal-dominant polycystic kidney disease (ADPKD) is a common genetic disease caused by mutations in the gene coding for polycystin-1 (PC1). PC1 can regulate STAT transcription factors by a novel, dual mechanism. STAT3 and STAT6 are aberrantly activated in renal cysts. Genetic and pharmacological approaches to inhibit STAT3 or STAT6 have led to promising results in ADPKD mouse models. Here, we review current findings that lead to a model of PC1 as a key regulator of STAT signaling in renal tubule cells. We discuss how PC1 may orchestrate appropriate epithelial responses to renal injury, and how this system may lead to aberrant STAT activation in ADPKD thereby causing inappropriate activation of tissue repair programs that culminate in renal cyst growth and fibrosis.
Collapse
Affiliation(s)
- Thomas Weimbs
- Department of Molecular, Cellular, and Developmental Biology; and Neuroscience Research Institute; University of California, Santa Barbara; Santa Barbara, CA USA
| | | | | |
Collapse
|