1
|
Anand A, Shenoy V, Sutherland RW, Platt C, Hildebrandt CC. Novel CHRNA3 variants identified in a patient with bladder dysfunction, dysautonomia, and gastrointestinal dysmotility. Am J Med Genet A 2024; 194:e63526. [PMID: 38192228 DOI: 10.1002/ajmg.a.63526] [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: 10/16/2023] [Revised: 12/14/2023] [Accepted: 12/18/2023] [Indexed: 01/10/2024]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are estimated to be responsible for 20%-50% of congenital anomalies and are also a leading etiology of early-onset renal disease. Primary CAKUT are caused by genetic factors that impair proper in-utero genitourinary tract development and secondary CAKUT result from the influence of environmental factors. The CHRNA3 gene, which encodes the Alpha-3 subunit of the nicotinic acetylcholine receptor, is hypothesized to be associated with Megacystis-microcolon-intestinal hyperperistalsis syndrome. More recently, pathogenic variants in CHRNA3 have been identified in individuals with CAKUT as well as individuals with panautonomic failure. Here we present a patient with neurogenic bladder, vesicoureteral reflux, mydriasis, and gastrointestinal dysmotility found to have novel compound heterozygous variants in CHRNA3. These findings support the consideration of CHRNA3 disruption in the differential for CAKUT with dysautonomia and gastrointestinal dysmotility.
Collapse
Affiliation(s)
- Asha Anand
- Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Vivek Shenoy
- UNC School of Medicine, Chapel Hill, North Carolina, USA
- UNC Department of Pediatrics, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | - Richard W Sutherland
- UNC School of Medicine, Chapel Hill, North Carolina, USA
- UNC Department of Urology, UNC School of Medicine, Chapel Hill, North Carolina, USA
| | | | - Clara C Hildebrandt
- UNC School of Medicine, Chapel Hill, North Carolina, USA
- UNC Department of Pediatrics, UNC School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
2
|
Thomson E, Tran M, Robevska G, Ayers K, van der Bergen J, Gopalakrishnan Bhaskaran P, Haan E, Cereghini S, Vash-Margita A, Margetts M, Hensley A, Nguyen Q, Sinclair A, Koopman P, Pelosi E. Functional genomics analysis identifies loss of HNF1B function as a cause of Mayer-Rokitansky-Küster-Hauser syndrome. Hum Mol Genet 2023; 32:1032-1047. [PMID: 36282544 PMCID: PMC9990990 DOI: 10.1093/hmg/ddac262] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/04/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome is a congenital condition characterized by aplasia or hypoplasia of the uterus and vagina in women with a 46,XX karyotype. This condition can occur as type I when isolated or as type II when associated with extragenital anomalies including kidney and skeletal abnormalities. The genetic basis of MRKH syndrome remains unexplained and several candidate genes have been proposed to play a role in its etiology, including HNF1B, LHX1 and WNT4. Here, we conducted a microarray analysis of 13 women affected by MRKH syndrome, resulting in the identification of chromosomal changes, including the deletion at 17q12, which contains both HNF1B and LHX1. We focused on HNF1B for further investigation due to its known association with, but unknown etiological role in, MRKH syndrome. We ablated Hnf1b specifically in the epithelium of the Müllerian ducts in mice and found that this caused hypoplastic development of the uterus, as well as kidney anomalies, closely mirroring the MRKH type II phenotype. Using single-cell RNA sequencing of uterine tissue in the Hnf1b-ablated embryos, we analyzed the molecules and pathways downstream of Hnf1b, revealing a dysregulation of processes associated with cell proliferation, migration and differentiation. Thus, we establish that loss of Hnf1b function leads to an MRKH phenotype and generate the first mouse model of MRKH syndrome type II. Our results support the investigation of HNF1B in clinical genetic settings of MRKH syndrome and shed new light on the molecular mechanisms underlying this poorly understood condition in women's reproductive health.
Collapse
Affiliation(s)
- Ella Thomson
- Centre for Clinical Research, The University of Queensland, Brisbane, QLD, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Minh Tran
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Gorjana Robevska
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | - Katie Ayers
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Jocelyn van der Bergen
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | | | - Eric Haan
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Silvia Cereghini
- Institut de Biologie Paris-Seine, Sorbonne Université, Paris, France
| | - Alla Vash-Margita
- Division of Pediatric and Adolescent Gynecology, Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
| | - Miranda Margetts
- Center for American Indian and Rural Health Equity, Montana State University, Bozeman, MT, USA
| | | | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - Andrew Sinclair
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | | | - Emanuele Pelosi
- To whom correspondence should be addressed. Tel: +61 7 3346 6073;
| |
Collapse
|
3
|
Warren EB, Briano JA, Ellegood J, DeYoung T, Lerch JP, Morrow EM. 17q12 deletion syndrome mouse model shows defects in craniofacial, brain and kidney development, and glucose homeostasis. Dis Model Mech 2022; 15:dmm049752. [PMID: 36373506 PMCID: PMC10655816 DOI: 10.1242/dmm.049752] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
17q12 deletion (17q12Del) syndrome is a copy number variant (CNV) disorder associated with neurodevelopmental disorders and renal cysts and diabetes syndrome (RCAD). Using CRISPR/Cas9 genome editing, we generated a mouse model of 17q12Del syndrome on both inbred (C57BL/6N) and outbred (CD-1) genetic backgrounds. On C57BL/6N, the 17q12Del mice had severe head development defects, potentially mediated by haploinsufficiency of Lhx1, a gene within the interval that controls head development. Phenotypes included brain malformations, particularly disruption of the telencephalon and craniofacial defects. On the CD-1 background, the 17q12Del mice survived to adulthood and showed milder craniofacial and brain abnormalities. We report postnatal brain defects using automated magnetic resonance imaging-based morphometry. In addition, we demonstrate renal and blood glucose abnormalities relevant to RCAD. On both genetic backgrounds, we found sex-specific presentations, with male 17q12Del mice exhibiting higher penetrance and more severe phenotypes. Results from these experiments pinpoint specific developmental defects and pathways that guide clinical studies and a mechanistic understanding of the human 17q12Del syndrome. This mouse mutant represents the first and only experimental model to date for the 17q12 CNV disorder. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Emily B. Warren
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI 02912, USA
| | - Juan A. Briano
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI 02912, USA
| | - Jacob Ellegood
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON M5T 3H7, Canada
| | - Taylor DeYoung
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON M5T 3H7, Canada
| | - Jason P. Lerch
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, ON M5T 3H7, Canada
- Wellcome Centre for Integrative Neuroimaging, The University of Oxford, Oxford OX3 9DU, UK
| | - Eric M. Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School of Brown University, Providence, RI 02912, USA
- Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI 02912, USA
| |
Collapse
|
4
|
Hnf1b renal expression directed by a distal enhancer responsive to Pax8. Sci Rep 2022; 12:19921. [PMID: 36402859 PMCID: PMC9675860 DOI: 10.1038/s41598-022-21171-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/23/2022] [Indexed: 11/21/2022] Open
Abstract
Xenopus provides a simple and efficient model system to study nephrogenesis and explore the mechanisms causing renal developmental defects in human. Hnf1b (hepatocyte nuclear factor 1 homeobox b), a gene whose mutations are the most commonly identified genetic cause of developmental kidney disease, is required for the acquisition of a proximo-intermediate nephron segment in Xenopus as well as in mouse. Genetic networks involved in Hnf1b expression during kidney development remain poorly understood. We decided to explore the transcriptional regulation of Hnf1b in the developing Xenopus pronephros and mammalian renal cells. Using phylogenetic footprinting, we identified an evolutionary conserved sequence (CNS1) located several kilobases (kb) upstream the Hnf1b transcription start and harboring epigenomic marks characteristics of a distal enhancer in embryonic and adult renal cells in mammals. By means of functional expression assays in Xenopus and mammalian renal cell lines we showed that CNS1 displays enhancer activity in renal tissue. Using CRISPR/cas9 editing in Xenopus tropicalis, we demonstrated the in vivo functional relevance of CNS1 in driving hnf1b expression in the pronephros. We further showed the importance of Pax8-CNS1 interaction for CNS1 enhancer activity allowing us to conclude that Hnf1b is a direct target of Pax8. Our work identified for the first time a Hnf1b renal specific enhancer and may open important perspectives into the diagnosis for congenital kidney anomalies in human, as well as modeling HNF1B-related diseases.
Collapse
|
5
|
Piedrafita A, Balayssac S, Casemayou A, Saulnier-Blache JS, Lucas A, Iacovoni JS, Breuil B, Chauveau D, Decramer S, Malet-Martino M, Schanstra JP, Faguer S. Hepatocyte nuclear factor-1β shapes the energetic homeostasis of kidney tubule cells. FASEB J 2021; 35:e21931. [PMID: 34653285 DOI: 10.1096/fj.202100782rr] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 08/28/2021] [Accepted: 09/02/2021] [Indexed: 12/17/2022]
Abstract
Energetic metabolism controls key steps of kidney development, homeostasis, and epithelial repair following acute kidney injury (AKI). Hepatocyte nuclear factor-1β (HNF-1β) is a master transcription factor that controls mitochondrial function in proximal tubule (PT) cells. Patients with HNF1B pathogenic variant display a wide range of kidney developmental abnormalities and progressive kidney fibrosis. Characterizing the metabolic changes in PT cells with HNF-1β deficiency may help to identify new targetable molecular hubs involved in HNF1B-related kidney phenotypes and AKI. Here, we combined 1 H-NMR-based metabolomic analysis in a murine PT cell line with CrispR/Cas9-induced Hnf1b invalidation (Hnf1b-/- ), clustering analysis, targeted metabolic assays, and datamining of published RNA-seq and ChIP-seq dataset to identify the role of HNF-1β in metabolism. Hnf1b-/- cells grown in normoxic conditions display intracellular ATP depletion, increased cytosolic lactate concentration, increased lipid droplet content, failure to use pyruvate for energetic purposes, increased levels of tricarboxylic acid (TCA) cycle intermediates and oxidized glutathione, and a reduction of TCA cycle byproducts, all features consistent with mitochondrial dysfunction and an irreversible switch toward glycolysis. Unsupervised clustering analysis showed that Hnf1b-/- cells mimic a hypoxic signature and that they cannot furthermore increase glycolysis-dependent energetic supply during hypoxic challenge. Metabolome analysis also showed alteration of phospholipid biosynthesis in Hnf1b-/- cells leading to the identification of Chka, the gene coding for choline kinase α, as a new putative target of HNF-1β. HNF-1β shapes the energetic metabolism of PT cells and HNF1B deficiency in patients could lead to a hypoxia-like metabolic state precluding further adaptation to ATP depletion following AKI.
Collapse
Affiliation(s)
- Alexis Piedrafita
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Balayssac
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France.,Laboratoire des Interaction Moléculaires et Réactivité Chimique et Photochimique (IMRCP), UMR 5623, Toulouse, France
| | - Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Jean-Sébastien Saulnier-Blache
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Alexandre Lucas
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Jason S Iacovoni
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Benjamin Breuil
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Service de Néphrologie, Médecine interne et Hypertension artérielle, Hôpital des Enfants, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| | - Myriam Malet-Martino
- Groupe de RMN Biomédicale, Laboratoire SPCMIB, UMR CNRS 5068, Université Paul Sabatier, Centre National de la Recherche Scientifique, Toulouse, France
| | - Joost P Schanstra
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, UMR 1297, Institut des Maladies Métaboliques et Cardiovasculaires, Hôpital Rangueil, Toulouse, France.,Université Paul Sabatier - Toulouse 3, Toulouse, France.,Département de Néphrologie et Transplantation d'Organes, Centre de Référence des Maladies Rénales Rares, Centre Hospitalier Universitaire de Toulouse, Toulouse, France
| |
Collapse
|
6
|
Kohl S, Habbig S, Weber LT, Liebau MC. Molecular causes of congenital anomalies of the kidney and urinary tract (CAKUT). Mol Cell Pediatr 2021; 8:2. [PMID: 33625646 PMCID: PMC7904997 DOI: 10.1186/s40348-021-00112-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 02/14/2021] [Indexed: 11/19/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) occur in 0.5–1/100 newborns and as a group they represent the most frequent cause for chronic kidney failure in children. CAKUT comprise clinically heterogeneous conditions, ranging from mild vesicoureteral reflux to kidney aplasia. Most forms of CAKUT share the pathophysiology of an impaired developmental interaction of the ureteric bud (UB) and the metanephric mesenchyme (MM). In most cases, CAKUT present as an isolated condition. They also may occur as a component in rare multi-organ syndromes. Many CAKUT probably have a multifactorial etiology. However, up to 20% of human patients and > 200 transgenic mouse models have a monogenic form of CAKUT, which has fueled our efforts to unravel molecular kidney (mal-)development. To date, genetic variants in more than 50 genes have been associated with (isolated) CAKUT in humans. In this short review, we will summarize typical imaging findings in patients with CAKUT and highlight recent mechanistic insight in the molecular pathogenesis of monogenic forms of CAKUT.
Collapse
Affiliation(s)
- Stefan Kohl
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.
| | - Sandra Habbig
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Lutz T Weber
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Max C Liebau
- Department of Pediatrics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany.,Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| |
Collapse
|
7
|
Chandra S, Srinivasan S, Batra J. Hepatocyte nuclear factor 1 beta: A perspective in cancer. Cancer Med 2021; 10:1791-1804. [PMID: 33580750 PMCID: PMC7940219 DOI: 10.1002/cam4.3676] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocyte nuclear factor 1 beta (HNF1 β/B) exists as a homeobox transcription factor having a vital role in the embryonic development of organs mainly liver, kidney and pancreas. Initially described as a gene causing maturity‐onset diabetes of the young (MODY), HNF1β expression deregulation and single nucleotide polymorphisms in HNF1β have now been associated with several tumours including endometrial, prostate, ovarian, hepatocellular, renal and colorectal cancers. Its function has been studied either as homodimer or heterodimer with HNF1α. In this review, the role of HNF1B in different cancers will be discussed along with the role of its splice variants, and its emerging role as a potential biomarker in cancer.
Collapse
Affiliation(s)
- Shubhra Chandra
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| | - Srilakshmi Srinivasan
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| | - Jyotsna Batra
- Institute of Health and Biomedical Innovation and School of Biomedical Sciences, Australian Prostate Cancer Research Centre-Queensland, Queensland University of Technology, Brisbane, QLD, Australia.,Translational Research Institute, Woolloongabba, QLD, Australia
| |
Collapse
|
8
|
Chan SC, Hajarnis SS, Vrba SM, Patel V, Igarashi P. Hepatocyte nuclear factor 1β suppresses canonical Wnt signaling through transcriptional repression of lymphoid enhancer-binding factor 1. J Biol Chem 2020; 295:17560-17572. [PMID: 33453998 DOI: 10.1074/jbc.ra120.015592] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/30/2020] [Indexed: 11/06/2022] Open
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is a tissue-specific transcription factor that is required for normal kidney development and renal epithelial differentiation. Mutations of HNF-1β produce congenital kidney abnormalities and inherited renal tubulopathies. Here, we show that ablation of HNF-1β in mIMCD3 renal epithelial cells results in activation of β-catenin and increased expression of lymphoid enhancer-binding factor 1 (LEF1), a downstream effector in the canonical Wnt signaling pathway. Increased expression and nuclear localization of LEF1 are also observed in cystic kidneys from Hnf1b mutant mice. Expression of dominant-negative mutant HNF-1β in mIMCD3 cells produces hyperresponsiveness to exogenous Wnt ligands, which is inhibited by siRNA-mediated knockdown of Lef1. WT HNF-1β binds to two evolutionarily conserved sites located 94 and 30 kb from the mouse Lef1 promoter. Ablation of HNF-1β decreases H3K27 trimethylation repressive marks and increases β-catenin occupancy at a site 4 kb upstream to Lef1. Mechanistically, WT HNF-1β recruits the polycomb-repressive complex 2 that catalyzes H3K27 trimethylation. Deletion of the β-catenin-binding domain of LEF1 in HNF-1β-deficient cells abolishes the increase in Lef1 transcription and decreases the expression of downstream Wnt target genes. The canonical Wnt target gene, Axin2, is also a direct transcriptional target of HNF-1β through binding to negative regulatory elements in the gene promoter. These findings demonstrate that HNF-1β regulates canonical Wnt target genes through long-range effects on histone methylation at Wnt enhancers and reveal a new mode of active transcriptional repression by HNF-1β.
Collapse
Affiliation(s)
- Siu Chiu Chan
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Sachin S Hajarnis
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sophia M Vrba
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Vishal Patel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter Igarashi
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
9
|
Mateus JC, Rivera C, O’Meara M, Valenzuela A, Lizcano F. Maturity-onset diabetes of the young type 5 a MULTISYSTEMIC disease: a CASE report of a novel mutation in the HNF1B gene and literature review. Clin Diabetes Endocrinol 2020; 6:16. [PMID: 32864159 PMCID: PMC7448977 DOI: 10.1186/s40842-020-00103-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diabetes mellitus with autosomal dominant inheritance, such as maturity-onset diabetes of the young (MODY), is a genetic form of diabetes mellitus. MODY is a type of monogenic diabetes mellitus in which multiple genetic variants may cause an alteration to the functioning of beta cells. The three most known forms of MODY are caused by modifications to the hnf4a, gck, and hnf1a genes. However, other MODY variants can cause multiple alterations in the embryonic development of the endoderm. This is the case in patients presenting with MODY5, who have a mutation of the hepatic nuclear factor 1B (hnf1b) gene. CASE PRESENTATION We present the clinical case of a 15 year-old patient with a family history of diabetes mellitus and a classical MODY type 5 (MODY5) phenotype involving the pancreas and kidney, with a novel, unreported mutation in the hnf1b gene. CONCLUSIONS MODY5 is characterised by a mutation in the hnf1b gene, which plays an important role in the development and function of multiple organs. It should be suspected in patients with unusual diabetes and multisystem involvement unrelated to diabetes. GRAPHICAL ABSTRACT
Collapse
Affiliation(s)
- Juan Camilo Mateus
- Endocrinology Fellowship, School of Medicine and Health Sciences, Rosary University – Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Carolina Rivera
- Department of Genetics, Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Miguel O’Meara
- Department of Endocrinology, Diabetes and Nutrition, Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Alex Valenzuela
- Department of Endocrinology, Diabetes and Nutrition, Fundacion Cardio-Infantil IC, Bogotá, Colombia
| | - Fernando Lizcano
- Department of Endocrinology, Diabetes and Nutrition, Fundacion Cardio-Infantil IC, Bogotá, Colombia
- Center of Biomedical Investigation Universidad de La Sabana, CIBUS, Chia, CU 250008 Colombia
| |
Collapse
|
10
|
Shu L, Hou G, Zhao H, Huang W, Song G, Ma H. Long non-coding RNA expression profiling following treatment with resveratrol to improve insulin resistance. Mol Med Rep 2020; 22:1303-1316. [PMID: 32627012 PMCID: PMC7339411 DOI: 10.3892/mmr.2020.11221] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 05/18/2020] [Indexed: 02/07/2023] Open
Abstract
Resveratrol (RSV) and long non-coding RNAs (lncRNAs) play a role in the treatment of diabetes; however, the mechanism by which resveratrol regulates insulin resistance via lncRNAs is currently unknown. The present study aimed to determine the lncRNA expression level profile in mice following resveratrol treatment to improve insulin resistance using high-throughput sequencing technology. C57BL/6J mice were fed a high-fat diet for 8 weeks to develop an insulin resistance model, followed by treatment with or without RSV for 6 weeks before high-throughput sequencing. Following RSV treatment, 28 and 30 lncRNAs were up- and downregulated, respectively; eight lncRNAs were randomly selected and evaluated using reverse transcription-quantitative PCR, which showed results consistent with the sequencing analysis. Pathway analysis demonstrated that the insulin signaling pathway enrichment score was the highest, and identified two lncRNAs, NONMMUT058999.2 and NONMMUT051901.2, consistent with the protein-encoding genes SOCS3 and G6PC, respectively. Similar expression level patterns were observed for SOCS3 and G6PC, suggesting that RSV improves insulin resistance by modulating lncRNAs. RSV decreased the expression levels of SOCS3, FOXO1, G6PC and PEPCK in mice. The same results were observed following knockdown of NONMMUT058999.2 in cells. The present study provides a new biomarker or intervention target for RSV in the treatment of diabetes, and a new perspective for understanding the hypoglycemic mechanism of RSV.
Collapse
Affiliation(s)
- Linyi Shu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Guangsen Hou
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Hang Zhao
- Endocrinology Department, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Wenli Huang
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Guangyao Song
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| | - Huijuan Ma
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei 050017, P.R. China
| |
Collapse
|
11
|
Shao A, Chan SC, Igarashi P. Role of transcription factor hepatocyte nuclear factor-1β in polycystic kidney disease. Cell Signal 2020; 71:109568. [PMID: 32068086 DOI: 10.1016/j.cellsig.2020.109568] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 02/09/2020] [Accepted: 02/12/2020] [Indexed: 02/07/2023]
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is a DNA-binding transcription factor that is essential for normal kidney development. Mutations of HNF1B in humans produce cystic kidney diseases, including renal cysts and diabetes, multicystic dysplastic kidneys, glomerulocystic kidney disease, and autosomal dominant tubulointerstitial kidney disease. Expression of HNF1B is reduced in cystic kidneys from humans with ADPKD, and HNF1B has been identified as a modifier gene in PKD. Genome-wide analysis of chromatin binding has revealed that HNF-1β directly regulates the expression of known PKD genes, such as PKHD1 and PKD2, as well as genes involved in PKD pathogenesis, including cAMP-dependent signaling, renal fibrosis, and Wnt signaling. In addition, a role of HNF-1β in regulating the expression of noncoding RNAs (microRNAs and long noncoding RNAs) has been identified. These findings indicate that HNF-1β regulates a transcriptional and post-transcriptional network that plays a central role in renal cystogenesis.
Collapse
Affiliation(s)
- Annie Shao
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Siu Chiu Chan
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Peter Igarashi
- Department of Medicine, University of Minnesota Medical School, Minneapolis, MN, USA.
| |
Collapse
|
12
|
Deng Q, Huang L, Liu J, Fang F, Liu Z, Zhang Y, Li F, Liao C. Prenatal diagnosis of submicroscopic chromosomal aberrations in fetuses with congenital cystic adenomatoid malformation by chromosomal microarray analysis. J Matern Fetal Neonatal Med 2019; 34:2623-2629. [PMID: 31581877 DOI: 10.1080/14767058.2019.1670793] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES To explore the copy number variations (CNVs) of fetal congenital cystic adenomatoid malformation (CCAM). METHODS Fetuses with CCAM were investigated by karyotypes and chromosomal microarray analysis (CMA). The cases were classified as isolated or CCAM with additional structural anomalies. The pregnancy outcome and neonatal prognosis were reported after the follow-up investigation. RESULTS The karyotypes of 43 fetuses were analyzed and no abnormal karyotype was detected. Thirty-seven cases were further tested using CMA. The CMA identified pathogenic CNVs in three fetuses with a pathogenic detection rate of 8.1%. Well-known microdeletion or microduplication syndromes, including RCAD syndrome, HNPP, and CMT1A were identified, among which HNPP and CMT1A were incidental findings. After excluding two incidental findings, there were no pathogenic CNVs in isolated CCAM. There were no significant differences in pathogenic CNVs between isolated CCAM and CCAM with additional structural anomalies (0%, 0/31 versus 16.7%, 1/6, p=.162). Nearly half of the patients (53.8%, 14/26) underwent surgery after birth with good postoperative recoveries while the remaining half patients were spontaneous regression or asymptomatic. CONCLUSIONS The results demonstrated the value of CMA in the prenatal diagnosis of CCAM. CCAM associated with other structural defects enhanced the frequency of pathogenic CNVs while isolated CCAM may not be associated with an increase in the prevalence of pathogenic CNVs. CCAMs have an overall good prognosis.
Collapse
Affiliation(s)
- Qiong Deng
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Lihua Huang
- Qingyuan Maternal and Child Health Hospital, Qingyuan, Guangdong, China
| | - Juan Liu
- Department of Obstetrics, Southern Medical University Affiliated Maternal and Child Health Hospital of Foshan, Foshan, Guangdong, China
| | - Fu Fang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Zequn Liu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Yongling Zhang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Fatao Li
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| | - Can Liao
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong, China
| |
Collapse
|
13
|
Al-Khawaga S, Mohammed I, Saraswathi S, Haris B, Hasnah R, Saeed A, Almabrazi H, Syed N, Jithesh P, El Awwa A, Khalifa A, AlKhalaf F, Petrovski G, Abdelalim EM, Hussain K. The clinical and genetic characteristics of permanent neonatal diabetes (PNDM) in the state of Qatar. Mol Genet Genomic Med 2019; 7:e00753. [PMID: 31441606 PMCID: PMC6785445 DOI: 10.1002/mgg3.753] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 04/04/2019] [Accepted: 04/27/2019] [Indexed: 02/06/2023] Open
Abstract
Background Neonatal diabetes mellitus (NDM) is a rare condition that occurs within the first six months of life. Permanent NDM (PNDM) is caused by mutations in specific genes that are known for their expression at early and/or late stages of pancreatic beta‐ cell development, and are either involved in beta‐cell survival, insulin processing, regulation, and release. The native population in Qatar continues to practice consanguineous marriages that lead to a high level of homozygosity. To our knowledge, there is no previous report on the genomics of NDM among the Qatari population. The aims of the current study are to identify patients with NDM diagnosed between 2001 and 2016, and examine their clinical and genetic characteristics. Methods To calculate the incidence of PNDM, all patients with PNDM diagnosed between 2001 and 2016 were compared to the total number of live births over the 16‐year‐period. Whole Genome Sequencing (WGS) was used to investigate the genetic etiology in the PNDM cohort. Results PNDM was diagnosed in nine (n = 9) patients with an estimated incidence rate of 1:22,938 live births among the indigenous Qatari. Seven different mutations in six genes (PTF1A, GCK, SLC2A2, EIF2AK3, INS, and HNF1B) were identified. In the majority of cases, the genetic etiology was part of a previously identified autosomal recessive disorder. Two novel de novo mutations were identified in INS and HNF1B. Conclusion Qatar has the second highest reported incidence of PNDM worldwide. A majority of PNDM cases present as rare familial autosomal recessive disorders. Pancreas associated transcription factor 1a (PTF1A) enhancer deletions are the most common cause of PNDM in Qatar, with only a few previous cases reported in the literature.
Collapse
Affiliation(s)
- Sara Al-Khawaga
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar.,Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Idris Mohammed
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Saras Saraswathi
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Basma Haris
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Reem Hasnah
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Amira Saeed
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | | | - Najeeb Syed
- Biomedical Informatics Division, Sidra Medicine, Doha, Qatar
| | - Puthen Jithesh
- Biomedical Informatics Division, Sidra Medicine, Doha, Qatar
| | - Ahmed El Awwa
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar.,Faculty of medicine, Alexandria University, Alexandria, Egypt
| | - Amal Khalifa
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Fawziya AlKhalaf
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Goran Petrovski
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| | - Essam M Abdelalim
- College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar.,Diabetes Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Khalid Hussain
- Division of Endocrinology, Department of Pediatric Medicine, Sidra Medicine, Doha, Qatar
| |
Collapse
|
14
|
Ferrè S, Igarashi P. New insights into the role of HNF-1β in kidney (patho)physiology. Pediatr Nephrol 2019; 34:1325-1335. [PMID: 29961928 PMCID: PMC6312759 DOI: 10.1007/s00467-018-3990-7] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 05/24/2018] [Accepted: 05/25/2018] [Indexed: 12/14/2022]
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is an essential transcription factor that regulates the development and function of epithelia in the kidney, liver, pancreas, and genitourinary tract. Humans who carry HNF1B mutations develop heterogeneous renal abnormalities, including multicystic dysplastic kidneys, glomerulocystic kidney disease, renal agenesis, renal hypoplasia, and renal interstitial fibrosis. In the embryonic kidney, HNF-1β is required for ureteric bud branching, initiation of nephrogenesis, and nephron segmentation. Ablation of mouse Hnf1b in nephron progenitors causes defective tubulogenesis, whereas later inactivation in elongating tubules leads to cyst formation due to downregulation of cystic disease genes, including Umod, Pkhd1, and Pkd2. In the adult kidney, HNF-1β controls the expression of genes required for intrarenal metabolism and solute transport by tubular epithelial cells. Tubular abnormalities observed in HNF-1β nephropathy include hyperuricemia with or without gout, hypokalemia, hypomagnesemia, and polyuria. Recent studies have identified novel post-transcriptional and post-translational regulatory mechanisms that control HNF-1β expression and activity, including the miRNA cluster miR17 ∼ 92 and the interacting proteins PCBD1 and zyxin. Further understanding of the molecular mechanisms upstream and downstream of HNF-1β may lead to the development of new therapeutic approaches in cystic kidney disease and other HNF1B-related renal diseases.
Collapse
Affiliation(s)
- Silvia Ferrè
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, Texas, USA,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter Igarashi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA. .,Department of Medicine, University of Minnesota Medical School, 420 Delaware St. SE, MMC 194, Minneapolis, MN, 55455, USA.
| |
Collapse
|
15
|
Ferrè S, Deng Y, Huen SC, Lu CY, Scherer PE, Igarashi P, Moe OW. Renal tubular cell spliced X-box binding protein 1 (Xbp1s) has a unique role in sepsis-induced acute kidney injury and inflammation. Kidney Int 2019; 96:1359-1373. [PMID: 31601454 DOI: 10.1016/j.kint.2019.06.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 06/04/2019] [Accepted: 06/27/2019] [Indexed: 12/15/2022]
Abstract
Sepsis is a systemic inflammatory state in response to infection, and concomitant acute kidney injury (AKI) increases mortality significantly. Endoplasmic reticulum stress is activated in many cell types upon microbial infection and modulates inflammation. The role of endoplasmic reticulum signaling in the kidney during septic AKI is unknown. Here we tested the role of the spliced X-box binding protein 1 (Xbp1s), a key component of the endoplasmic reticulum stress-activated pathways, in the renal response to sepsis in the lipopolysaccharide (LPS) model. Xbp1s was increased in the kidneys of mice treated with LPS but not in other models of AKI, or several chronic kidney disease models. The functional significance of Xbp1s induction was examined by genetic manipulation in renal tubules. Renal tubule-specific overexpression of Xbp1s caused severe tubule dilation and vacuolation with expression of the injury markers Kim1 and Ngal, the pro-inflammatory molecules interleukin-6 (Il6) and Toll-like receptor 4 (Tlr4), decreased kidney function and 50% mortality in five days. Renal tubule-specific genetic ablation of Xbp1 had no phenotype at baseline. However, after LPS, Xbp1 knockdown mice displayed lower renal NGAL, pro-apoptotic factor CHOP, serum creatinine levels, and a tendency towards lower Tlr4 compared to LPS-treated mice with intact Xbp1s. LPS treatment in Xbp1s-overexpressing mice caused a mild increase in NGAL and CHOP compared to LPS-treated mice without genetic Xbp1s overexpression. Thus, increased Xbp1s signaling in renal tubules is unique to sepsis-induced AKI and contributes to renal inflammation and injury. Inhibition of this pathway may be a potential portal to alleviate injury.
Collapse
Affiliation(s)
- Silvia Ferrè
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| | - Yingfeng Deng
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sarah C Huen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Y Lu
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Philipp E Scherer
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Touchstone Diabetes Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Peter Igarashi
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Orson W Moe
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Department of Physiology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
16
|
Sander V, Salleh L, Naylor RW, Schierding W, Sontam D, O’Sullivan JM, Davidson AJ. Transcriptional profiling of the zebrafish proximal tubule. Am J Physiol Renal Physiol 2019; 317:F478-F488. [DOI: 10.1152/ajprenal.00174.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The hepatocyte nuclear factor-1β (Hnf1b) transcription factor is a key regulator of kidney tubule formation and is associated with a syndrome of renal cysts and early onset diabetes. To further our understanding of Hnf1b in the developing zebrafish kidney, we performed RNA sequencing analysis of proximal tubules from hnf1b-deficient larvae. This analysis revealed an enrichment of gene transcripts encoding transporters of the solute carrier (SLC) superfamily, including multiple members of slc2 and slc5 glucose transporters. An investigation of expression of slc2a1a, slc2a2, and slc5a2 as well as a poorly studied glucose/mannose transporter encoded by slc5a9 revealed that these genes undergo dynamic spatiotemporal changes during tubule formation and maturation. A comparative analysis of zebrafish SLC genes with those expressed in mouse proximal tubules showed a substantial overlap at the level of gene families, indicating a high degree of functional conservation between zebrafish and mammalian proximal tubules. Taken together, our findings are consistent with a role for Hnf1b as a critical determinant of proximal tubule transport function by acting upstream of a large number of SLC genes and validate the zebrafish as a physiologically relevant model of the mammalian proximal tubule.
Collapse
Affiliation(s)
- Veronika Sander
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Liam Salleh
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Richard W. Naylor
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | | | - Dharani Sontam
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Alan J. Davidson
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| |
Collapse
|
17
|
Loeschenberger B, Niess L, Würzner R, Schwelberger H, Eder IE, Puhr M, Guenther J, Troppmair J, Rudnicki M, Neuwirt H. Calcineurin inhibitor-induced complement system activation via ERK1/2 signalling is inhibited by SOCS-3 in human renal tubule cells. Eur J Immunol 2017; 48:330-343. [PMID: 29143318 DOI: 10.1002/eji.201747135] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/12/2017] [Accepted: 11/03/2017] [Indexed: 12/22/2022]
Abstract
One factor that significantly contributes to renal allograft loss is chronic calcineurin inhibitor (CNI) nephrotoxicity (CIN). Among other factors, the complement (C-) system has been proposed to be involved CIN development. Hence, we investigated the impact of CNIs on intracellular signalling and the effects on the C-system in human renal tubule cells. In a qPCR array, CNI treatment upregulated C-factors and downregulated SOCS-3 and the complement inhibitors CD46 and CD55. Additionally, ERK1/-2 was required for these regulations. Following knock-down and overexpression of SOCS-3, we found that SOCS-3 inhibits ERK1/-2 signalling. Finally, we assessed terminal complement complex formation, cell viability and apoptosis. Terminal complement complex formation was induced by CNIs. Cell viability was significantly decreased, whereas apoptosis was increased. Both effects were reversed under complement component-depleted conditions. In vivo, increased ERK1/-2 phosphorylation and SOCS-3 downregulation were observed at the time of transplantation in renal allograft patients who developed a progressive decline of renal function in the follow-up compared to stable patients. The progressive cohort also had lower total C3 levels, suggesting higher complement activity at baseline. In conclusion, our data suggest that SOCS-3 inhibits CNI-induced ERK1/-2 signalling, thereby blunting the negative control of C-system activation.
Collapse
Affiliation(s)
| | - Lea Niess
- Department of Internal Medicine IV, Innsbruck Medical University, Innsbruck, Austria
| | - Reinhard Würzner
- Division of Hygiene and Medical Microbiology, Innsbruck Medical University, Innsbruck, Austria
| | - Hubert Schwelberger
- Molecular Biology Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Iris E Eder
- Department of Urology, Division of Experimental Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Martin Puhr
- Department of Urology, Division of Experimental Urology, Innsbruck Medical University, Innsbruck, Austria
| | - Julia Guenther
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant- and Thoracic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Jakob Troppmair
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant- and Thoracic Surgery, Innsbruck Medical University, Innsbruck, Austria
| | - Michael Rudnicki
- Department of Internal Medicine IV, Innsbruck Medical University, Innsbruck, Austria
| | - Hannes Neuwirt
- Department of Internal Medicine IV, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
18
|
Casemayou A, Fournel A, Bagattin A, Schanstra J, Belliere J, Decramer S, Marsal D, Gillet M, Chassaing N, Huart A, Pontoglio M, Knauf C, Bascands JL, Chauveau D, Faguer S. Hepatocyte Nuclear Factor-1 β Controls Mitochondrial Respiration in Renal Tubular Cells. J Am Soc Nephrol 2017; 28:3205-3217. [PMID: 28739648 DOI: 10.1681/asn.2016050508] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 05/18/2017] [Indexed: 12/19/2022] Open
Abstract
AKI is a frequent condition that involves renal microcirculation impairment, infiltration of inflammatory cells with local production of proinflammatory cytokines, and subsequent epithelial disorders and mitochondrial dysfunction. Peroxisome proliferator-activated receptor γ coactivator 1-α (PPARGC1A), a coactivator of the transcription factor PPAR-γ that controls mitochondrial biogenesis and function, has a pivotal role in the early dysfunction of the proximal tubule and the subsequent renal repair. Here, we evaluated the potential role of hepatocyte nuclear factor-1β (HNF-1β) in regulating PPARGC1A expression in AKI. In mice, endotoxin injection to induce AKI also induced early and transient inflammation and PPARGC1A inhibition, which overlapped with downregulation of the HNF-1β transcriptional network. In vitro, exposure of proximal tubule cells to the inflammatory cytokines IFN-γ and TNF-α led to inhibition of HNF-1β transcriptional activity. Moreover, inhibition of HNF-1β significantly reduced PPARGC1A expression and altered mitochondrial morphology and respiration in proximal tubule cells. Chromatin immunoprecipitation assays and PCR analysis confirmed HNF-1β binding to the Ppargc1a promoter in mouse kidneys. We also demonstrated downregulation of renal PPARGC1A expression in a patient with an HNF1B germinal mutation. Thus, we propose that HNF-1β links extracellular inflammatory signals to mitochondrial dysfunction during AKI partly via PPARGC1A signaling. Our findings further strengthen the view of HNF1B-related nephropathy as a mitochondrial disorder in adulthood.
Collapse
Affiliation(s)
- Audrey Casemayou
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Audren Fournel
- University Toulouse III Paul-Sabatier, Toulouse, France.,Institut National de la Santé et de la Recherche Médicale U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan-BP3028, 31024 Toulouse Cedex 3
| | - Alessia Bagattin
- Laboratoire d'Expression Génique, Développement et Maladies, Département Développement, Reproduction et Cancer, Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| | - Joost Schanstra
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Julie Belliere
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Stéphane Decramer
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology, Internal Medicine and Hypertension, Center for Rare Renal Diseases, Children' Hospital, University Hospital of Toulouse, Toulouse, France
| | - Dimitri Marsal
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Marion Gillet
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France
| | - Nicolas Chassaing
- Department of Medical Genetics, Hôpital Purpan, University Hospital of Toulouse, Toulouse, France; and
| | - Antoine Huart
- Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Marco Pontoglio
- Laboratoire d'Expression Génique, Développement et Maladies, Département Développement, Reproduction et Cancer, Institut National de la Santé et de la Recherche Médicale U1016, Institut Cochin, Paris, France
| | - Claude Knauf
- University Toulouse III Paul-Sabatier, Toulouse, France.,Institut National de la Santé et de la Recherche Médicale U1220, Institut de Recherche en Santé Digestive (IRSD), CHU Purpan-BP3028, 31024 Toulouse Cedex 3
| | - Jean-Loup Bascands
- Institut National de la Santé et de la Recherche Médicale, U1188, DéTROI (Diabète Athérothrombose Thérapies Réunion Océan Indien), University of La Réunion
| | - Dominique Chauveau
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France.,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| | - Stanislas Faguer
- Institut National de la Santé et de la Recherche Médicale, U1048, Institut of Cardiovascular and Metabolic Disease, Toulouse, France; .,University Toulouse III Paul-Sabatier, Toulouse, France.,Department of Nephrology and Organ Transplantation, Center for Rare Renal Diseases, University Hospital of Toulouse, Toulouse, France
| |
Collapse
|
19
|
Aboudehen K, Noureddine L, Cobo-Stark P, Avdulov S, Farahani S, Gearhart MD, Bichet DG, Pontoglio M, Patel V, Igarashi P. Hepatocyte Nuclear Factor-1 β Regulates Urinary Concentration and Response to Hypertonicity. J Am Soc Nephrol 2017; 28:2887-2900. [PMID: 28507058 DOI: 10.1681/asn.2016101095] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 04/16/2017] [Indexed: 12/18/2022] Open
Abstract
The transcription factor hepatocyte nuclear factor-1β (HNF-1β) is essential for normal kidney development and function. Inactivation of HNF-1β in mouse kidney tubules leads to early-onset cyst formation and postnatal lethality. Here, we used Pkhd1/Cre mice to delete HNF-1β specifically in renal collecting ducts (CDs). CD-specific HNF-1β mutant mice survived long term and developed slowly progressive cystic kidney disease, renal fibrosis, and hydronephrosis. Compared with wild-type littermates, HNF-1β mutant mice exhibited polyuria and polydipsia. Before the development of significant renal structural abnormalities, mutant mice exhibited low urine osmolality at baseline and after water restriction and administration of desmopressin. However, mutant and wild-type mice had similar plasma vasopressin and solute excretion levels. HNF-1β mutant kidneys showed increased expression of aquaporin-2 mRNA but mislocalized expression of aquaporin-2 protein in the cytoplasm of CD cells. Mutant kidneys also had decreased expression of the UT-A urea transporter and collectrin, which is involved in apical membrane vesicle trafficking. Treatment of HNF-1β mutant mIMCD3 cells with hypertonic NaCl inhibited the induction of osmoregulated genes, including Nr1h4, which encodes the transcription factor FXR that is required for maximal urinary concentration. Chromatin immunoprecipitation and sequencing experiments revealed HNF-1β binding to the Nr1h4 promoter in wild-type kidneys, and immunoblot analysis revealed downregulated expression of FXR in HNF-1β mutant kidneys. These findings reveal a novel role of HNF-1β in osmoregulation and identify multiple mechanisms, whereby mutations of HNF-1β produce defects in urinary concentration.
Collapse
Affiliation(s)
- Karam Aboudehen
- Departments of Medicine and.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lama Noureddine
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.,Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Patricia Cobo-Stark
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | - Micah D Gearhart
- Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota
| | - Daniel G Bichet
- Departments of Medicine and.,Molecular and Integrative Physiology, Université de Montréal, Montreal, Quebec, Canada; and
| | - Marco Pontoglio
- Department of Development, Reproduction and Cancer, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016/Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris-Descartes, Paris, France
| | - Vishal Patel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peter Igarashi
- Departments of Medicine and .,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
20
|
Patitucci C, Couchy G, Bagattin A, Cañeque T, de Reyniès A, Scoazec JY, Rodriguez R, Pontoglio M, Zucman-Rossi J, Pende M, Panasyuk G. Hepatocyte nuclear factor 1α suppresses steatosis-associated liver cancer by inhibiting PPARγ transcription. J Clin Invest 2017; 127:1873-1888. [PMID: 28394260 DOI: 10.1172/jci90327] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 02/16/2017] [Indexed: 12/26/2022] Open
Abstract
Worldwide epidemics of metabolic diseases, including liver steatosis, are associated with an increased frequency of malignancies, showing the highest positive correlation for liver cancer. The heterogeneity of liver cancer represents a clinical challenge. In liver, the transcription factor PPARγ promotes metabolic adaptations of lipogenesis and aerobic glycolysis under the control of Akt2 activity, but the role of PPARγ in liver tumorigenesis is unknown. Here we have combined preclinical mouse models of liver cancer and genetic studies of a human liver biopsy atlas with the aim of identifying putative therapeutic targets in the context of liver steatosis and cancer. We have revealed a protumoral interaction of Akt2 signaling with hepatocyte nuclear factor 1α (HNF1α) and PPARγ, transcription factors that are master regulators of hepatocyte and adipocyte differentiation, respectively. Akt2 phosphorylates and inhibits HNF1α, thus relieving the suppression of hepatic PPARγ expression and promoting tumorigenesis. Finally, we observed that pharmacological inhibition of PPARγ is therapeutically effective in a preclinical murine model of steatosis-associated liver cancer. Taken together, our studies in humans and mice reveal that Akt2 controls hepatic tumorigenesis through crosstalk between HNF1α and PPARγ.
Collapse
|
21
|
Matsui A, Fujimoto J, Ishikawa K, Ito E, Goshima N, Watanabe S, Semba K. Hepatocyte nuclear factor 1 beta induces transformation and epithelial-to-mesenchymal transition. FEBS Lett 2016; 590:1211-21. [PMID: 27001343 DOI: 10.1002/1873-3468.12147] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/11/2016] [Accepted: 03/16/2016] [Indexed: 12/31/2022]
Abstract
Gene amplification can be a cause of cancer, and driver oncogenes have been often identified in amplified regions. However, comprehensive analysis of other genes coamplified with an oncogene is rarely performed. We focused on the 17q12-21 amplicon, which contains ERBB2. We established a screening system for oncogenic activity with the NMuMG epithelial cell line. We identified a homeobox gene, HNF1B, as a novel cooperative transforming gene. HNF1B induced cancerous phenotypes, which were enhanced by the coexpression of ERBB2, and induced epithelial-to-mesenchymal transition and invasive phenotypes. These results suggest that HNF1B is a novel oncogene that can work cooperatively with ERBB2.
Collapse
Affiliation(s)
- Atsuka Matsui
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| | - Jiro Fujimoto
- Japan Biological Informatics Consortium (JBiC), Tokyo, Japan.,Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Kosuke Ishikawa
- Japan Biological Informatics Consortium (JBiC), Tokyo, Japan.,Research Institute for Science and Engineering, Waseda University, Tokyo, Japan
| | - Emi Ito
- Division of Gene Expression Analysis, Translational Research Center, Fukushima Medical University, Japan
| | - Naoki Goshima
- Division of Transcriptome Analysis, Translational Research Center, Fukushima Medical University, Japan.,Quantitative Proteomics Team, Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, Japan
| | - Shinya Watanabe
- Division of Gene Expression Analysis, Translational Research Center, Fukushima Medical University, Japan
| | - Kentaro Semba
- Department of Life Science and Medical Bioscience, School of Advanced Science and Engineering, Waseda University, Tokyo, Japan.,Division of Gene Function Analysis, Translational Research Center, Fukushima Medical University, Japan
| |
Collapse
|
22
|
Aboudehen K, Kim MS, Mitsche M, Garland K, Anderson N, Noureddine L, Pontoglio M, Patel V, Xie Y, DeBose-Boyd R, Igarashi P. Transcription Factor Hepatocyte Nuclear Factor-1β Regulates Renal Cholesterol Metabolism. J Am Soc Nephrol 2015; 27:2408-21. [PMID: 26712526 DOI: 10.1681/asn.2015060607] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 11/11/2015] [Indexed: 12/16/2022] Open
Abstract
HNF-1β is a tissue-specific transcription factor that is expressed in the kidney and other epithelial organs. Humans with mutations in HNF-1β develop kidney cysts, and HNF-1β regulates the transcription of several cystic disease genes. However, the complete spectrum of HNF-1β-regulated genes and pathways is not known. Here, using chromatin immunoprecipitation/next generation sequencing and gene expression profiling, we identified 1545 protein-coding genes that are directly regulated by HNF-1β in murine kidney epithelial cells. Pathway analysis predicted that HNF-1β regulates cholesterol metabolism. Expression of dominant negative mutant HNF-1β or kidney-specific inactivation of HNF-1β decreased the expression of genes that are essential for cholesterol synthesis, including sterol regulatory element binding factor 2 (Srebf2) and 3-hydroxy-3-methylglutaryl-CoA reductase (Hmgcr). HNF-1β mutant cells also expressed lower levels of cholesterol biosynthetic intermediates and had a lower rate of cholesterol synthesis than control cells. Additionally, depletion of cholesterol in the culture medium mitigated the inhibitory effects of mutant HNF-1β on the proteins encoded by Srebf2 and Hmgcr, and HNF-1β directly controlled the renal epithelial expression of proprotein convertase subtilisin-like kexin type 9, a key regulator of cholesterol uptake. These findings reveal a novel role of HNF-1β in a transcriptional network that regulates intrarenal cholesterol metabolism.
Collapse
Affiliation(s)
- Karam Aboudehen
- Departments of Internal Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota; and
| | | | | | | | | | | | - Marco Pontoglio
- Department of Development, Reproduction and Cancer, National Institute of Health and Medical Research (INSERM) U1016, The National Center for Scientific Research (CNRS) Joint Research Unit (UMR) 8104, University of Paris Descartes, Institut Cochin, Paris, France
| | | | | | - Russell DeBose-Boyd
- Molecular Genetics, and Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peter Igarashi
- Departments of Internal Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota; and Pediatrics and
| |
Collapse
|
23
|
Mandai M, Amano Y, Yamaguchi K, Matsumura N, Baba T, Konishi I. Ovarian clear cell carcinoma meets metabolism; HNF-1β confers survival benefits through the Warburg effect and ROS reduction. Oncotarget 2015; 6:30704-14. [PMID: 26375553 PMCID: PMC4741562 DOI: 10.18632/oncotarget.5228] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 08/10/2015] [Indexed: 12/21/2022] Open
Abstract
Ovarian clear cell carcinoma (OCCC) constitutes one of the subtypes of ovarian cancers, but it has unique clinical, histological and biological characteristics, one of which is chemo-resistance. It is also known to develop from endometriotic cyst, a benign ovarian tumor, at relatively high frequency. Recently, it is becoming well known that most of OCCCs express HNF1β, a transcription factor, which is closely associated with the development of liver, pancreas and kidney, as well as occurrence of familial forms of type 2 diabetes. Expression of HNF1β is now regarded as a hallmark of this tumor. Nevertheless, exact biological function of this gene in OCCC has not been clarified. We have shown in previous studies that microenvironment in endometriotic cysts contains severe oxidative stress and OCCC develops under such stressful environment as stress-resistant tumor, which may lead to chemo-resistance. We also showed that increased expression of HNF1β facilitates glucose uptake and glycolysis, which is known as Warburg effect. In the previous issue of this journal, by using comprehensive metabolome analysis, we report that HNF1β actually reduces and protects themselves from internal oxidative stress by dramatically changing cellular metabolism. In this article, we review the relevance and significance of cancer-specific metabolism and how they are associated with biological characteristics of OCCC via expression of HNF1β, along with future clinical implications of targeting cancer-specific metabolism.
Collapse
Affiliation(s)
- Masaki Mandai
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Obstetrics and Gynecology, Kinki University Faculty of Medicine, Osaka-Sayama, Japan
| | - Yasuaki Amano
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ken Yamaguchi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tsukasa Baba
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ikuo Konishi
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
24
|
Hajarnis SS, Patel V, Aboudehen K, Attanasio M, Cobo-Stark P, Pontoglio M, Igarashi P. Transcription Factor Hepatocyte Nuclear Factor-1β (HNF-1β) Regulates MicroRNA-200 Expression through a Long Noncoding RNA. J Biol Chem 2015; 290:24793-805. [PMID: 26292219 DOI: 10.1074/jbc.m115.670646] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Indexed: 12/31/2022] Open
Abstract
The transcription factor hepatocyte nuclear factor-1β (HNF-1β) regulates tissue-specific gene expression in the kidney and other epithelial organs. Mutations of HNF-1β produce kidney cysts, and previous studies have shown that HNF-1β regulates the transcription of cystic disease genes, including Pkd2 and Pkhd1. Here, we combined chromatin immunoprecipitation and next-generation sequencing (ChIP-Seq) with microarray analysis to identify microRNAs (miRNAs) that are directly regulated by HNF-1β in renal epithelial cells. These studies identified members of the epithelial-specific miR-200 family (miR-200b/200a/429) as novel transcriptional targets of HNF-1β. HNF-1β binds to two evolutionarily conserved sites located 28 kb upstream to miR-200b. Luciferase reporter assays showed that the HNF-1β binding sites were located within a promoter that was active in renal epithelial cells. Mutations of the HNF-1β binding sites abolished promoter activity. RT-PCR analysis revealed that a long noncoding RNA (lncRNA) is transcribed from the promoter and encodes the miR-200 cluster. Inhibition of the lncRNA with siRNAs decreased the levels of miR-200 but did not affect expression of the Ttll10 host gene. The expression of the lncRNA and miR-200 was decreased in kidneys from HNF-1β knock-out mice and renal epithelial cells expressing dominant-negative mutant HNF-1β. The expression of miR-200 targets, Zeb2 and Pkd1, was increased in HNF-1β knock-out kidneys and in cells expressing mutant HNF-1β. Overexpression of miR-200 decreased the expression of Zeb2 and Pkd1 in HNF-1β mutant cells. These studies reveal a novel pathway whereby HNF-1β directly contributes to the control of miRNAs that are involved in epithelial-mesenchymal transition and cystic kidney disease.
Collapse
Affiliation(s)
| | | | | | | | | | - Marco Pontoglio
- Départment de Génétique et Développement, INSERM U1016, CNRS UMR 8104, Université Paris-Descartes. Institut Cochin, 75014 Paris, France
| | - Peter Igarashi
- From the Departments of Internal Medicine and Pediatrics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390 and
| |
Collapse
|
25
|
Musetti C, Quaglia M, Mellone S, Pagani A, Fusco I, Monzani A, Giordano M, Stratta P. Chronic renal failure of unknown origin is caused by HNF1B mutations in 9% of adult patients: a single centre cohort analysis. Nephrology (Carlton) 2014; 19:202-9. [PMID: 24387224 DOI: 10.1111/nep.12199] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/23/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND HNF1B gene mutations might be an underdiagnosed cause of nephropathy in adult patients mainly because of their pleomorphic clinical presentations. As most studies are based on paediatric populations, it is difficult to assess the likelihood of finding HNF1B mutations in adult patients and consequently define clinical settings in which genetic analysis is indicated. The aim of this study was the search for mutations in the HNF1B gene in a cohort of unrelated adult patients with nephropathy of unknown aetiology. METHODS Patients were tested for the HNF1B gene if they had chronic kidney disease of unknown origin and renal structure abnormalities (RSA) or a positive family history of nephropathy. The HNF1B coding sequence and intron-exon boundaries were analysed by direct sequencing. The search for gene deletions was performed by Multiple Ligation Probe Analysis (MLPA). RESULTS Heterozygous mutations were identified in 6 out of 67 screened patients (9.0%) and included two whole gene deletions, one nonsense (p.Gln136Stop), two missense (p.Gly76Cys and p.Ala314Thr) mutations and a frameshift microdeletion (c.384_390 delCATGCAG), the latter two (c.384_390 del and p.Ala314Thr) not ever being reported to date. Mean age of the mutated patients at screening was 48.5 years with a M/F ratio of 2/4. The clinical manifestations of affected patients were extremely pleomorphic, including several urological and extra-renal manifestations. CONCLUSIONS Mutations of HNF1B could explain chronic kidney disease in up to 9% of adult patients with a nephropathy of unknown aetiology and RSA: therefore an HNF1B mutation analysis should be considered in this group of patients.
Collapse
Affiliation(s)
- Claudio Musetti
- Department of Translational Medicine, Nephrology and Transplantation, Hospital 'Maggiore della Carità', Novara, NO, Italy
| | | | | | | | | | | | | | | |
Collapse
|
26
|
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
|
27
|
Williams SS, Cobo-Stark P, Hajarnis S, Aboudehen K, Shao X, Richardson JA, Patel V, Igarashi P. Tissue-specific regulation of the mouse Pkhd1 (ARPKD) gene promoter. Am J Physiol Renal Physiol 2014; 307:F356-68. [PMID: 24899057 DOI: 10.1152/ajprenal.00422.2013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Autosomal recessive polycystic kidney disease, an inherited disorder characterized by the formation of cysts in renal collecting ducts and biliary dysgenesis, is caused by mutations of the polycystic kidney and hepatic disease 1 (PKHD1) gene. Expression of PKHD1 is tissue specific and developmentally regulated. Here, we show that a 2.0-kb genomic fragment containing the proximal promoter of mouse Pkhd1 directs tissue-specific expression of a lacZ reporter gene in transgenic mice. LacZ is expressed in renal collecting ducts beginning during embryonic development but is not expressed in extrarenal tissues. The Pkhd1 promoter contains a binding site for the transcription factor hepatocyte nuclear factor (HNF)-1β, which is required for activity in transfected cells. Mutation of the HNF-1β-binding site abolishes the expression of the lacZ reporter gene in renal collecting ducts. Transgenes containing the 2.0-kb promoter and 2.7 kb of additional genomic sequence extending downstream to the second exon are expressed in the kidney, intrahepatic bile ducts, and male reproductive tract. This pattern overlaps with the endogenous expression of Pkhd1 and coincides with sites of expression of HNF-1β. We conclude that the proximal 2.0-kb promoter is sufficient for tissue-specific expression of Pkhd1 in renal collecting ducts in vivo and that HNF-1β is required for Pkhd1 promoter activity in collecting ducts. Additional genomic sequences located from exons 1-2 or elsewhere in the gene locus are required for expression in extrarenal tissues.
Collapse
Affiliation(s)
- Scott S Williams
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Patricia Cobo-Stark
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sachin Hajarnis
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Karam Aboudehen
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Xinli Shao
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - James A Richardson
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; and Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vishal Patel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Peter Igarashi
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas;
| |
Collapse
|
28
|
Naylor RW, Davidson AJ. Hnf1beta and nephron segmentation. Pediatr Nephrol 2014; 29:659-64. [PMID: 24190171 PMCID: PMC3944118 DOI: 10.1007/s00467-013-2662-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Revised: 10/03/2013] [Accepted: 10/09/2013] [Indexed: 01/03/2023]
Abstract
The nephron is the functional unit that executes the homeostatic roles of the kidney in vertebrates. Critical to this function is the physical arrangement of the glomerular blood filter attached to a tubular epithelium that is subdivided into specialized proximal and distal segments. During embryogenesis, nephron progenitors undergo a mesenchymal-epithelial transition (MET) and adopt different segment-specific cell fates along the proximo-distal axis of the nephron. The molecular basis of how these segments arise remains largely unknown. Recent studies using the zebrafish have identified the Hnf1beta transcription factor (Hnf1b) as a major regulator of tubular segmentation. In Hnf1b-deficient zebrafish embryos, nephron progenitors fail to adopt the proximo-distal segmentation pattern of the nephron, yet still undergo MET. This observation suggests that the functional segmentation of renal tubular epithelial cells is independent of pathways that induce their epithelialization. Here we review this new role of Hnf1b for nephron segmentation during zebrafish and mouse kidney development.
Collapse
|
29
|
Ablation of proximal tubular suppressor of cytokine signaling 3 enhances tubular cell cycling and modifies macrophage phenotype during acute kidney injury. Kidney Int 2014; 85:1357-68. [PMID: 24402091 DOI: 10.1038/ki.2013.525] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 09/18/2013] [Accepted: 10/17/2013] [Indexed: 01/23/2023]
Abstract
Suppressor of cytokine signaling 3 (SOCS-3) is an important intracellular negative regulator of several signaling pathways. We found that SOCS-3 is highly expressed in renal proximal tubules during acute kidney injury. To test the impact of this, conditional proximal tubular knockout mice (SOCS-3(sglt2Δ/sglt2Δ)) were created. These mice had better kidney function than their wild-type counterparts in aristolochic acid nephropathy and after ischemia/reperfusion injury. Kidneys of these knockout mice showed significantly more proximal tubular cell proliferation during the repair phase. A direct effect of SOCS-3 on tubular cell cycling was demonstrated by in vitro experiments showing a JAK/STAT pathway-dependent antimitotic effect of SOCS-3. Furthermore, acute damaged kidneys of the knockout mice contained increased numbers of F4/80(+) cells. Phenotypic analysis of these F4/80(+) cells indicated a polarization from classically activated to alternatively activated macrophages. In vitro, SOCS-3-overexpressing renal epithelial cells directly induced classical activation in cocultured macrophages, supporting the observed in vivo phenomenon. Thus, upregulation of SOCS-3 in stressed proximal tubules plays an important role during acute kidney injury by inhibition of reparative proliferation and by modulation of the macrophage phenotype. Antagonizing SOCS-3 could have therapeutic potential for acute kidney injury.
Collapse
|
30
|
Abstract
Polycystic diseases affect approximately 1/1000 and are important causes of kidney failure. No therapies presently are in clinical practice that can prevent disease progression. Multiple mouse models have been produced for the genetic forms of the disease that most commonly affect humans. In this report, we review recent progress in the field and describe some of the outstanding challenges.
Collapse
Affiliation(s)
- Luis Fernando Menezes
- Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Building 10 Room 8D46, 10 Center Drive, Bethesda, MD 20892
| | - Gregory George Germino
- Kidney Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Building 10 Room 8D46, 10 Center Drive, Bethesda, MD 20892
| |
Collapse
|
31
|
Chauveau D, Faguer S, Bandin F, Guigonis V, Chassaing N, Decramer S. HNF1B : paradigme d’un gène du développement et émergence inattendue d’une nouvelle maladie génétique rénale. Nephrol Ther 2013; 9:393-7. [DOI: 10.1016/j.nephro.2013.05.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Revised: 05/13/2013] [Accepted: 05/13/2013] [Indexed: 11/30/2022]
|
32
|
miR-17~92 miRNA cluster promotes kidney cyst growth in polycystic kidney disease. Proc Natl Acad Sci U S A 2013; 110:10765-70. [PMID: 23759744 DOI: 10.1073/pnas.1301693110] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Polycystic kidney disease (PKD), the most common genetic cause of chronic kidney failure, is characterized by the presence of numerous, progressively enlarging fluid-filled cysts in the renal parenchyma. The cysts arise from renal tubules and are lined by abnormally functioning and hyperproliferative epithelial cells. Despite recent progress, no Food and Drug Administration-approved therapy is available to retard cyst growth. MicroRNAs (miRNAs) are short noncoding RNAs that inhibit posttranscriptional gene expression. Dysregulated miRNA expression is observed in PKD, but whether miRNAs are directly involved in kidney cyst formation and growth is not known. Here, we show that miR-17∼92, an oncogenic miRNA cluster, is up-regulated in mouse models of PKD. Kidney-specific transgenic overexpression of miR-17∼92 produces kidney cysts in mice. Conversely, kidney-specific inactivation of miR-17∼92 in a mouse model of PKD retards kidney cyst growth, improves renal function, and prolongs survival. miR-17∼92 may mediate these effects by promoting proliferation and through posttranscriptional repression of PKD genes Pkd1, Pkd2, and hepatocyte nuclear factor-1β. These studies demonstrate a pathogenic role of miRNAs in mouse models of PKD and identify miR-17∼92 as a therapeutic target in PKD. Our results also provide a unique hypothesis for disease progression in PKD involving miRNAs and regulation of PKD gene dosage.
Collapse
|
33
|
White CA, Nicola NA. SOCS3: An essential physiological inhibitor of signaling by interleukin-6 and G-CSF family cytokines. JAKSTAT 2013; 2:e25045. [PMID: 24416642 PMCID: PMC3876435 DOI: 10.4161/jkst.25045] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Accepted: 05/15/2013] [Indexed: 12/21/2022] Open
Abstract
SOCS3 is an inducible negative feedback inhibitor of cytokine signaling. Conditional deletion of SOCS3 in mice using the Cre-lox system has now been applied to a range of cell types in the steady-state and under inflammatory, pathogenic, or tumorigenic stress, with the resulting phenotypes demonstrating the effects of SOCS3 in physiological and disease contexts. Together with recent structural and biochemical studies on the mechanisms of SOCS3 binding to cytokine receptors and associated kinases, we now have a better understanding of the non-redundant roles of SOCS3 in the inhibition of cytokine signaling via the receptors gp130, G-CSFR, leptinR, and IL-12Rβ. This review discusses the known functional activities of SOCS3 in fertility and development, inflammation, innate and adaptive immunity, and malignancy as determined by genetic studies in mice.
Collapse
Affiliation(s)
- Christine A White
- Walter and Eliza Hall Institute of Medical Research; Parkville, VIC Australia ; Department of Medical Biology; University of Melbourne; Parkville, VIC Australia
| | - Nicos A Nicola
- Walter and Eliza Hall Institute of Medical Research; Parkville, VIC Australia ; Department of Medical Biology; University of Melbourne; Parkville, VIC Australia
| |
Collapse
|
34
|
Faguer S, Mayeur N, Casemayou A, Pageaud AL, Courtellemont C, Cartery C, Fournie GJ, Schanstra JP, Tack I, Bascands JL, Chauveau D. Hnf-1β transcription factor is an early hif-1α-independent marker of epithelial hypoxia and controls renal repair. PLoS One 2013; 8:e63585. [PMID: 23704921 PMCID: PMC3660442 DOI: 10.1371/journal.pone.0063585] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Accepted: 04/08/2013] [Indexed: 01/19/2023] Open
Abstract
Epithelial repair following acute kidney injury (AKI) requires epithelial-mesenchyme-epithelial cycling associated with transient re-expression of genes normally expressed during kidney development as well as activation of growth factors and cytokine-induced signaling. In normal kidney, the Hnf-1β transcription factor drives nephrogenesis, tubulogenesis and epithelial homeostasis through the regulation of epithelial planar cell polarity and expression of developmental or tubular segment-specific genes. In a mouse model of ischemic AKI induced by a 2-hours hemorrhagic shock, we show that expression of this factor is tightly regulated in the early phase of renal repair with a biphasic expression profile (early down-regulation followed by transient over-expression). These changes are associated to tubular epithelial differentiation as assessed by KSP-cadherin and megalin-cubilin endocytic complex expression analysis. In addition, early decrease in Hnf1b expression is associated with the transient over-expression of one of its main target genes, the suppressor of cytokine signaling Socs3, which has been shown essential for renal repair. In vitro, hypoxia induced early up-regulation of Hnf-1β from 1 to 24 hours, independently of the hypoxia-inducible factor Hif-1α. When prolonged, hypoxia induced Hnf-1β down-regulation while normoxia led to Hnf-1β normalization. Last, Hnf-1β down-regulation using RNA interference in HK-2 cells led to phenotype switch from an epithelial to a mesenchyme state. Taken together, we showed that Hnf-1β may drive recovery from ischemic AKI by regulating both the expression of genes important for homeostasis control during organ repair and the state of epithelial cell differentiation.
Collapse
MESH Headings
- Acute Kidney Injury/genetics
- Acute Kidney Injury/pathology
- Acute Kidney Injury/physiopathology
- Animals
- Biomarkers/metabolism
- Cell Hypoxia/drug effects
- Cell Hypoxia/genetics
- Cell Line
- Disease Models, Animal
- Endocytosis/drug effects
- Epithelial Cells/drug effects
- Epithelial Cells/metabolism
- Epithelial Cells/pathology
- Female
- Gene Expression Regulation/drug effects
- Hepatocyte Nuclear Factor 1-beta/antagonists & inhibitors
- Hepatocyte Nuclear Factor 1-beta/genetics
- Hepatocyte Nuclear Factor 1-beta/metabolism
- Humans
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Kidney/drug effects
- Kidney/metabolism
- Kidney/pathology
- Kidney/physiopathology
- Kidney Tubules, Proximal/drug effects
- Kidney Tubules, Proximal/metabolism
- Kidney Tubules, Proximal/pathology
- Kidney Tubules, Proximal/physiopathology
- Mice
- Mice, Inbred C57BL
- Prolyl-Hydroxylase Inhibitors/pharmacology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Shock, Hemorrhagic/complications
- Shock, Hemorrhagic/pathology
- Shock, Hemorrhagic/physiopathology
- Wound Healing/drug effects
Collapse
Affiliation(s)
- Stanislas Faguer
- Département de Néphrologie et Transplantation d'organes, CHU Rangueil, Toulouse, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Choi YH, McNally BT, Igarashi P. Zyxin regulates migration of renal epithelial cells through activation of hepatocyte nuclear factor-1β. Am J Physiol Renal Physiol 2013; 305:F100-10. [PMID: 23657850 DOI: 10.1152/ajprenal.00582.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is an epithelial tissue-specific transcription factor that regulates gene expression in the kidney, liver, pancreas, intestine, and other organs. Mutations of HNF-1β in humans produce renal cysts and congenital kidney anomalies. Here, we identify the LIM-domain protein zyxin as a novel binding partner of HNF-1β in renal epithelial cells. Zyxin shuttles to the nucleus where it colocalizes with HNF-1β. Immunoprecipitation of zyxin in leptomycin B-treated cells results in coprecipitation of HNF-1β. The protein interaction requires the second LIM domain of zyxin and two distinct domains of HNF-1β. Overexpression of zyxin stimulates the transcriptional activity of HNF-1β, whereas small interfering RNA silencing of zyxin inhibits HNF-1β-dependent transcription. Epidermal growth factor (EGF) induces translocation of zyxin into the nucleus and stimulates HNF-1β-dependent promoter activity. The EGF-mediated nuclear translocation of zyxin requires activation of Akt. Expression of dominant-negative mutant HNF-1β, knockdown of zyxin, or inhibition of Akt inhibits EGF-stimulated cell migration. These findings reveal a novel pathway by which extracellular signals are transmitted to the nucleus to regulate the activity of a transcription factor that is essential for renal epithelial differentiation.
Collapse
Affiliation(s)
- Yun-Hee Choi
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | |
Collapse
|
36
|
Kornfeld JW, Baitzel C, Könner AC, Nicholls HT, Vogt MC, Herrmanns K, Scheja L, Haumaitre C, Wolf AM, Knippschild U, Seibler J, Cereghini S, Heeren J, Stoffel M, Brüning JC. Obesity-induced overexpression of miR-802 impairs glucose metabolism through silencing of Hnf1b. Nature 2013; 494:111-5. [PMID: 23389544 DOI: 10.1038/nature11793] [Citation(s) in RCA: 264] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Accepted: 11/15/2012] [Indexed: 12/16/2022]
Abstract
Insulin resistance represents a hallmark during the development of type 2 diabetes mellitus and in the pathogenesis of obesity-associated disturbances of glucose and lipid metabolism. MicroRNA (miRNA)-dependent post-transcriptional gene silencing has been recognized recently to control gene expression in disease development and progression, including that of insulin-resistant type 2 diabetes. The deregulation of miRNAs miR-143 (ref. 4), miR-181 (ref. 5), and miR-103 and miR-107 (ref. 6) alters hepatic insulin sensitivity. Here we report that the expression of miR-802 is increased in the liver of two obese mouse models and obese human subjects. Inducible transgenic overexpression of miR-802 in mice causes impaired glucose tolerance and attenuates insulin sensitivity, whereas reduction of miR-802 expression improves glucose tolerance and insulin action. We identify Hnf1b (also known as Tcf2) as a target of miR-802-dependent silencing, and show that short hairpin RNA (shRNA)-mediated reduction of Hnf1b in liver causes glucose intolerance, impairs insulin signalling and promotes hepatic gluconeogenesis. In turn, hepatic overexpression of Hnf1b improves insulin sensitivity in Lepr(db/db) mice. Thus, this study defines a critical role for deregulated expression of miR-802 in the development of obesity-associated impairment of glucose metabolism through targeting of Hnf1b, and assigns Hnf1b an unexpected role in the control of hepatic insulin sensitivity.
Collapse
Affiliation(s)
- Jan-Wilhelm Kornfeld
- Max-Planck-Institute for Neurological Research, Gleueler Strasse 50a, 50931 Cologne, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Naylor RW, Przepiorski A, Ren Q, Yu J, Davidson AJ. HNF1β is essential for nephron segmentation during nephrogenesis. J Am Soc Nephrol 2012; 24:77-87. [PMID: 23160512 DOI: 10.1681/asn.2012070756] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Nephrons comprise a blood filter and an epithelial tubule that is subdivided into proximal and distal segments, but what directs this patterning during kidney organogenesis is not well understood. Using zebrafish, we found that the HNF1β paralogues hnf1ba and hnf1bb, which encode homeodomain transcription factors, are essential for normal segmentation of nephrons. Embryos deficient in hnf1ba and hnf1bb did not express proximal and distal segment markers, yet still developed an epithelial tubule. Initiating hnf1ba/b expression required Pax2a and Pax8, but hnf1ba/b-deficient embryos did not exhibit the expected downregulation of pax2a and pax8 at later stages of development, suggesting complex regulatory loops involving these molecules. Embryos deficient in hnf1ba/b also did not express the irx3b transcription factor, which is responsible for differentiation of the first distal tubule segment. Reciprocally, embryos deficient in irx3b exhibited downregulation of hnf1ba/b transcripts in the distal early segment, suggesting a segment-specific regulatory circuit. Deficiency of hnf1ba/b also led to ectopic expansion of podocytes into the proximal tubule domain. Epistasis experiments showed that the formation of podocytes required wt1a, which encodes the Wilms' tumor suppressor-1 transcription factor, and rbpj, which encodes a mediator of canonical Notch signaling, downstream or parallel to hnf1ba/b. Taken together, these results suggest that Hnf1β factors are essential for normal segmentation of nephrons during kidney organogenesis.
Collapse
Affiliation(s)
- Richard W Naylor
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland 1142, New Zealand
| | | | | | | | | |
Collapse
|
38
|
Ogata K, Shimamura Y, Hamada K, Hisa M, Bun M, Okada N, Inoue K, Taniguchi Y, Ishihara M, Kagawa T, Horino T, Fujimoto S, Terada Y. Upregulation of HNF-1β during experimental acute kidney injury plays a crucial role in renal tubule regeneration. Am J Physiol Renal Physiol 2012; 303:F689-99. [PMID: 22759397 DOI: 10.1152/ajprenal.00086.2012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Hepatocyte nuclear factor-1β (HNF-1β) is a transcription factor expressed in the kidney, liver, pancreas, and other organs. Mutations of HNF-1β cause maturity-onset diabetes of the young type 5 (MODY5). The aims of this study were to investigate the functional roles of the HNF-1β/suppressor of cytokine signaling-3 (SOCS-3) pathway in tubule damage after acute kidney injury (AKI) both in vivo and in vitro and to examine the effect of HNF-1β on renal tubule formation. To clarify the significance of the HNF-1β/SOCS-3 pathway in AKI, we used a rat ischemia/reperfusion (I/R) AKI model and cultured renal tubular cells (NRK-52E cells). Western blot analysis showed that HNF-1β and polycystic kidney disease 2 (PKD2) expressions were increased at 3-12 h and 12-24 h after I/R, respectively. The expression level of SOCS-3 was decreased at 3-48 h. Immunohistological examination revealed that expression of HNF-1β was increased in proximal tubules. Overexpression of HNF-1β resulted in decreased SOCS-3 expression, activation of signal transducer and activator of transcription 3 (STAT3) and Erk, and increased [(3)H]thymidine uptake in the presence of hepatocyte growth factor. Furthermore, tubule formation in three-dimensional gels was inhibited by dominant-negative HNF-1β. Our study shows that HNF-1β is upregulated after AKI in proximal tubular cells and that HNF-1β controls cellular proliferation and tubule formation by regulating SOCS-3 expression and STAT3/Erk activation. Therefore, the current study unravels the physiological and pathological significance of the HNF-1β pathway in AKI.
Collapse
Affiliation(s)
- Koji Ogata
- Dept. of Endocrinology, Metabolism and Nephrology, Kochi Medical School, Kochi University, Kohasu, Oko-cho, Nankoku, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Choi YH, Suzuki A, Hajarnis S, Ma Z, Chapin HC, Caplan MJ, Pontoglio M, Somlo S, Igarashi P. Polycystin-2 and phosphodiesterase 4C are components of a ciliary A-kinase anchoring protein complex that is disrupted in cystic kidney diseases. Proc Natl Acad Sci U S A 2011; 108:10679-84. [PMID: 21670265 PMCID: PMC3127890 DOI: 10.1073/pnas.1016214108] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Polycystic kidney disease (PKD) is a genetic disorder that is characterized by cyst formation in kidney tubules. PKD arises from abnormalities of the primary cilium, a sensory organelle located on the cell surface. Here, we show that the primary cilium of renal epithelial cells contains a protein complex comprising adenylyl cyclase 5/6 (AC5/6), A-kinase anchoring protein 150 (AKAP150), and protein kinase A. Loss of primary cilia caused by deletion of Kif3a results in activation of AC5 and increased cAMP levels. Polycystin-2 (PC2), a ciliary calcium channel that is mutated in human PKD, interacts with AC5/6 through its C terminus. Deletion of PC2 increases cAMP levels, which can be corrected by reexpression of wild-type PC2 but not by a mutant lacking calcium channel activity. Phosphodiesterase 4C (PDE4C), which catabolizes cAMP, is also located in renal primary cilia and interacts with the AKAP150 complex. Expression of PDE4C is regulated by the transcription factor hepatocyte nuclear factor-1β (HNF-1β), mutations of which produce kidney cysts. PDE4C is down-regulated and cAMP levels are increased in HNF-1β mutant kidney cells and mice. Collectively, these findings identify PC2 and PDE4C as unique components of an AKAP complex in primary cilia and reveal a common mechanism for dysregulation of cAMP signaling in cystic kidney diseases arising from different gene mutations.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marco Pontoglio
- Institut National de la Santé et de la Recherche Médicale U567, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris-Descartes, Institut Cochin, 75014 Paris, France
| | - Stefan Somlo
- Internal Medicine and Genetics, Yale University School of Medicine, New Haven, CT 06520; and
| | - Peter Igarashi
- Departments of Internal Medicine and
- Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|
40
|
Anglesio MS, George J, Kulbe H, Friedlander M, Rischin D, Lemech C, Power J, Coward J, Cowin PA, House CM, Chakravarty P, Gorringe KL, Campbell IG, Australian Ovarian Cancer Study Group, Okamoto A, Birrer MJ, Huntsman DG, de Fazio A, Kalloger SE, Balkwill F, Gilks CB, Bowtell DD. IL6-STAT3-HIF signaling and therapeutic response to the angiogenesis inhibitor sunitinib in ovarian clear cell cancer. Clin Cancer Res 2011; 17:2538-48. [PMID: 21343371 DOI: 10.1158/1078-0432.ccr-10-3314] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Collaborators] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
PURPOSE Ovarian clear cell adenocarcinoma (OCCA) is an uncommon histotype that is generally refractory to platinum-based chemotherapy. We analyze here the most comprehensive gene expression and copy number data sets, to date, to identify potential therapeutic targets of OCCA. EXPERIMENTAL DESIGN Gene expression and DNA copy number were carried out using primary human OCCA tumor samples, and findings were confirmed by immunohistochemistry on tissue microarrays. Circulating interleukin (IL) 6 levels were measured in serum from patients with OCCA or high-grade serous cancers and related to progression-free and overall survival. Two patients were treated with sunitinib, and their therapeutic responses were measured clinically and by positron emission tomography. RESULTS We find specific overexpression of the IL6-STAT3-HIF (interleukin 6-signal transducer and activator of transcription 3-hypoxia induced factor) pathway in OCCA tumors compared with high-grade serous cancers. Expression of PTHLH and high levels of circulating IL6 in OCCA patients may explain the frequent occurrence of hypercalcemia of malignancy and thromboembolic events in OCCA. We describe amplification of several receptor tyrosine kinases, most notably MET, suggesting other potential therapeutic targets. We report sustained clinical and functional imaging responses in two OCCA patients with chemotherapy-resistant disease who were treated with sunitinib, thus showing significant parallels with renal clear cell cancer. CONCLUSIONS Our findings highlight important therapeutic targets in OCCA, suggest that more extensive clinical trials with sunitinib in OCCA are warranted, and provide significant impetus to the growing realization that OCCA is molecularly and clinically distinct to other forms of ovarian cancer.
Collapse
Collaborators
D Bowtell, A Green, P Webb, A deFazio, D Gertig,
Collapse
|
41
|
Szponar A, Yusenko MV, Kuiper R, van Kessel AG, Kovacs G. Genomic profiling of papillary renal cell tumours identifies small regions of DNA alterations: a possible role of HNF1B in tumour development. Histopathology 2011; 58:934-43. [DOI: 10.1111/j.1365-2559.2011.03795.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
42
|
Ferrè S, Veenstra GJC, Bouwmeester R, Hoenderop JG, Bindels RJ. HNF-1B specifically regulates the transcription of the γa-subunit of the Na+/K+-ATPase. Biochem Biophys Res Commun 2011; 404:284-90. [DOI: 10.1016/j.bbrc.2010.11.108] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Accepted: 11/24/2010] [Indexed: 10/18/2022]
|
43
|
Kim D, Kim SH, Cho SH, Shin K, Kim S. SOCS3 suppresses the expression of IL-4 cytokine by inhibiting the phosphorylation of c-Jun through the ERK signaling pathway in rat mast cell line RBL-2H3. Mol Immunol 2010; 48:776-81. [PMID: 21168220 DOI: 10.1016/j.molimm.2010.11.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 10/19/2010] [Accepted: 11/06/2010] [Indexed: 11/27/2022]
Abstract
SOCS3 is well known to negatively regulate various cytokine-mediated signaling responses, but its direct role in the expression of specific cytokines has not been clearly elucidated. To understand the role of SOCS3 in the expression of IL-4, one of the key Th2 cytokines, RBL-2H3 cells (a rat mast cell line) were engineered to express SOCS3 constitutively at a high level or at a lower level using shRNA. In RBL-2H3 cells stably expressing SOCS3, the RNA and protein levels of IL-4 were significantly decreased, while it was opposite in RBL-2H3 cells containing shRNA for SOCS3. Overexpression of SOCS3 was found to reduce the level of calcium ionophore-induced phosphorylation of ERK1/2 and c-Jun transcription factor. Consistent with this data, knockdown of SOCS3 increased the level of phosphorylated ERK1 and ERK2. Taken together, SOCS3 appears to play an important role as a negative feedback inhibitor in the expression of IL-4 by inhibiting serine phosphorylation of c-Jun via the ERK signaling pathway.
Collapse
Affiliation(s)
- Donghyun Kim
- School of Biological Sciences, Seoul National University, Gwanak-gu, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
44
|
Matsumura N, Mandai M, Okamoto T, Yamaguchi K, Yamamura S, Oura T, Baba T, Hamanishi J, Kang HS, Matsui S, Mori S, Murphy SK, Konishi I. Sorafenib efficacy in ovarian clear cell carcinoma revealed by transcriptome profiling. Cancer Sci 2010; 101:2658-63. [PMID: 21040214 PMCID: PMC11159119 DOI: 10.1111/j.1349-7006.2010.01736.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The purpose of this study was to investigate a new modality of therapy against ovarian clear cell carcinoma (OCCC), a chemoresistant subtype of ovarian cancer. Microarray datasets of ovarian cancer cell lines and cancer tissues were analyzed using bioinformatic tools. The gene expression profile of OCCC was similar to that of renal cell carcinoma (RCC). This similarity was at least partially due to hepatocyte nuclear factor 1 pathway activation common to both malignancies. In addition, oncogenic pathway alterations were characteristic of OCCC including hypoxia inducible factor 1 alpha subunit and relatively high Ras activities. Therefore, we predicted that the multi-kinase inhibitor sorafenib, which is approved for RCC and suppresses Ras activity, would also be effective against OCCC. Orally administered sorafenib (40 mg/kg per day) significantly inhibited tumor growth in nude mice when it was given after inoculation with the OCCC cell line RMG-2 (P = 0.002). Furthermore, sorafenib significantly reduced tumor size when it was administered to established RMG-2 tumors (P = 0.0002), while intraperitoneal injection of cisplatin (5 mg/kg per week) did not. In conclusion, the prominent anti-tumor effect of sorafenib against OCCC indicates that sorafenib is a promising candidate drug and supports the need for clinical trials using sorafenib against OCCC. This report demonstrates a method to utilize genome-wide information to facilitate translational research for treatments against less common subtypes of cancers.
Collapse
Affiliation(s)
- Noriomi Matsumura
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Kim M, O'Brien LE, Kwon SH, Mostov KE. STAT1 is required for redifferentiation during Madin-Darby canine kidney tubulogenesis. Mol Biol Cell 2010; 21:3926-33. [PMID: 20861313 PMCID: PMC2982126 DOI: 10.1091/mbc.e10-02-0112] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Signal transducers and activators of transcription (STAT)1 is the key to the sequential control of Madin-Darby canine kidney tubulogenesis. Loss of STAT1 prevents redifferentiation. Constitutively active STAT1 is sufficient to restore cord formation but not mature lumens. These data suggest that STAT1 is necessary for the redifferentiation phase of tubulogenesis and that mature lumenogenesis requires a distinct signal. Tubule formation in vitro using Madin-Darby canine kidney (MDCK) epithelial cells consists mainly of two processes. First, the cells undergo a partial epithelial–mesenchymal transition (pEMT), losing polarity and migrating. Second, the cells redifferentiate, forming cords and then tubules with continuous lumens. We have shown previously that extracellular signal-regulated kinase activation is required for pEMT. However, the mechanism of how the pEMT phase is turned off and the redifferentiation phase is initiated is largely unknown. To address the central question of the sequential control of these two phases, we used MDCK cells grown as cysts and treated with hepatocyte growth factor to model tubulogenesis. We show that signal transducer and activator of transcription (STAT)1 controls the sequential progression from the pEMT phase to the redifferentiation phase. Loss of STAT1 prevents redifferentiation. Constitutively active STAT1 allows redifferentiation to occur even when cells are otherwise prevented from progressing beyond the pEMT phase by exogenous activation of Raf. Moreover, tyrosine phosphorylation defective STAT1 partially restored cord formation in such cells, suggesting that STAT1 functions in part as nonnuclear protein mediating signal transduction in this process. Constitutively active or inactive forms of STAT1 did not promote lumen maturation, suggesting this requires a distinct signal.
Collapse
Affiliation(s)
- Minji Kim
- Department of Anatomy, University of California-San Francisco, San Francisco, CA 94158, USA
| | | | | | | |
Collapse
|
46
|
Hahn H. Genetics of kidney development: pathogenesis of renal anomalies. KOREAN JOURNAL OF PEDIATRICS 2010; 53:729-34. [PMID: 21189947 PMCID: PMC3004483 DOI: 10.3345/kjp.2010.53.7.729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2010] [Revised: 06/08/2010] [Accepted: 06/14/2010] [Indexed: 11/30/2022]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) account for more than 50% of abdominal masses found in neonates and involve about 0.5% of all pregnancies. CAKUT has a major role in renal failure, and increasing evidence suggests that certain abnormalities predispose to the development of hypertension and cardiovascular disease in adulthood. To understand the pathogenesis of human renal anomalies, understanding the development of kidney is important. Diverse anomalies of the kidney corresponding to defects at a particular stage of development have been documented recently; however, more research is required to understand the molecular networks underlying kidney development, and such an investigation will provide a clue to the therapeutic intervention for CAKUT.
Collapse
Affiliation(s)
- Hyewon Hahn
- Department of Pediatrics, Eulji University School of Medicine, Seoul, Korea
| |
Collapse
|
47
|
Molecular anatomy of the kidney: what have we learned from gene expression and functional genomics? Pediatr Nephrol 2010; 25:1005-16. [PMID: 20049614 PMCID: PMC3189493 DOI: 10.1007/s00467-009-1392-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2009] [Revised: 09/15/2009] [Accepted: 09/18/2009] [Indexed: 12/15/2022]
Abstract
The discipline of paediatric nephrology encompasses the congenital nephritic syndromes, renal dysplasias, neonatal renal tumours, early onset cystic disease, tubulopathies and vesicoureteric reflux, all of which arise due to defects in normal kidney development. Indeed, congenital anomalies of the kidney and urinary tract (CAKUT) represent 20-30% of prenatal anomalies, occurring in 1 in 500 births. Developmental biologists have studied the anatomical and morphogenetic processes involved in kidney development for the last five decades. However, with the advent of transgenic mice, the sequencing of the genome, improvements in mutation detection and the advent of functional genomics, our understanding of the molecular basis of kidney development has grown significantly. Here we discuss how the advent of new genetic and genomics approaches has added to our understanding of kidney development and paediatric renal disease, as well as identifying areas in which we are still lacking knowledge.
Collapse
|
48
|
O'Toole JF, Liu Y, Davis EE, Westlake CJ, Attanasio M, Otto EA, Seelow D, Nurnberg G, Becker C, Nuutinen M, Kärppä M, Ignatius J, Uusimaa J, Pakanen S, Jaakkola E, van den Heuvel LP, Fehrenbach H, Wiggins R, Goyal M, Zhou W, Wolf MTF, Wise E, Helou J, Allen SJ, Murga-Zamalloa CA, Ashraf S, Chaki M, Heeringa S, Chernin G, Hoskins BE, Chaib H, Gleeson J, Kusakabe T, Suzuki T, Isaac RE, Quarmby LM, Tennant B, Fujioka H, Tuominen H, Hassinen I, Lohi H, van Houten JL, Rotig A, Sayer JA, Rolinski B, Freisinger P, Madhavan SM, Herzer M, Madignier F, Prokisch H, Nurnberg P, Jackson PK, Jackson P, Khanna H, Katsanis N, Hildebrandt F. Individuals with mutations in XPNPEP3, which encodes a mitochondrial protein, develop a nephronophthisis-like nephropathy. J Clin Invest 2010; 120:791-802. [PMID: 20179356 DOI: 10.1172/jci40076] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Accepted: 01/06/2010] [Indexed: 01/06/2023] Open
Abstract
The autosomal recessive kidney disease nephronophthisis (NPHP) constitutes the most frequent genetic cause of terminal renal failure in the first 3 decades of life. Ten causative genes (NPHP1-NPHP9 and NPHP11), whose products localize to the primary cilia-centrosome complex, support the unifying concept that cystic kidney diseases are "ciliopathies". Using genome-wide homozygosity mapping, we report here what we believe to be a new locus (NPHP-like 1 [NPHPL1]) for an NPHP-like nephropathy. In 2 families with an NPHP-like phenotype, we detected homozygous frameshift and splice-site mutations, respectively, in the X-prolyl aminopeptidase 3 (XPNPEP3) gene. In contrast to all known NPHP proteins, XPNPEP3 localizes to mitochondria of renal cells. However, in vivo analyses also revealed a likely cilia-related function; suppression of zebrafish xpnpep3 phenocopied the developmental phenotypes of ciliopathy morphants, and this effect was rescued by human XPNPEP3 that was devoid of a mitochondrial localization signal. Consistent with a role for XPNPEP3 in ciliary function, several ciliary cystogenic proteins were found to be XPNPEP3 substrates, for which resistance to N-terminal proline cleavage resulted in attenuated protein function in vivo in zebrafish. Our data highlight an emerging link between mitochondria and ciliary dysfunction, and suggest that further understanding the enzymatic activity and substrates of XPNPEP3 will illuminate novel cystogenic pathways.
Collapse
Affiliation(s)
- John F O'Toole
- Department of Pediatrics, University of Michigan, Ann Arbor, 48109-5646, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Verdeguer F, Le Corre S, Fischer E, Callens C, Garbay S, Doyen A, Igarashi P, Terzi F, Pontoglio M. A mitotic transcriptional switch in polycystic kidney disease. Nat Med 2009; 16:106-10. [PMID: 19966811 DOI: 10.1038/nm.2068] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2009] [Accepted: 11/05/2009] [Indexed: 02/07/2023]
Abstract
Hepatocyte nuclear factor-1beta (HNF-1beta) is a transcription factor required for the expression of several renal cystic genes and whose prenatal deletion leads to polycystic kidney disease (PKD). We show here that inactivation of Hnf1b from postnatal day 10 onward does not elicit cystic dilations in tubules after their proliferative morphogenetic elongation is over. Cystogenic resistance is intrinsically linked to the quiescent state of cells. In fact, when Hnf1b deficient quiescent cells are forced to proliferate by an ischemia-reperfusion injury, they give rise to cysts, owing to loss of oriented cell division. Remarkably, in quiescent cells, the transcription of crucial cystogenic target genes is maintained even in the absence of HNF-1beta. However, their expression is lost as soon as cells proliferate and the chromatin of target genes acquires heterochromatin marks. These results unveil a previously undescribed aspect of gene regulation. It is well established that transcription is shut off during the mitotic condensation of chromatin. We propose that transcription factors such as HNF-1beta might be involved in reprogramming gene expression after transcriptional silencing is induced by mitotic chromatin condensation. Notably, HNF-1beta remains associated with the mitotically condensed chromosomal barrels. This association suggests that HNF-1beta is a bookmarking factor that is necessary for reopening the chromatin of target genes after mitotic silencing.
Collapse
Affiliation(s)
- Francisco Verdeguer
- Gene Expression, Development and Disease Laboratory, Developmental Biology Department, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Kiu H, Greenhalgh CJ, Thaus A, Hilton DJ, Nicola NA, Alexander WS, Roberts AW. Regulation of multiple cytokine signalling pathways by SOCS3 is independent of SOCS2. Growth Factors 2009; 27:384-93. [PMID: 19919527 PMCID: PMC3412171 DOI: 10.3109/08977190903210954] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Suppressor of cytokine signalling (SOCS) 3 is an essential regulator of cytokine signalling, and in turn its expression is tightly regulated. Data from overexpression studies in cell lines suggest that SOCS2 regulates SOCS3 protein degradation, by forming a molecular bridge to an E3 ubiquitin-ligase complex. Whether this regulation is relevant in primary cells is unknown. In this study, we utilized Socs2( - / - ) mice to examine the role of SOCS2 in modulating SOCS3 expression and degradation, and its impact on interleukin-2 (IL-2) and IL-6 signalling in primary haemopoietic cells. Both biochemical and biological analyses demonstrated unperturbed SOCS3 expression and cytokine signalling in the absence of SOCS2. Our results suggest that SOCS2 is not a physiological regulator of SOCS3 expression and action in primary haemopoietic cells.
Collapse
Affiliation(s)
- Hiu Kiu
- Division of Cancer and Haematology, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, 3050, Australia
| | | | | | | | | | | | | |
Collapse
|