151
|
Mukherjee M, deRiso J, Otterpohl K, Ratnayake I, Kota D, Ahrenkiel P, Chandrasekar I, Surendran K. Endogenous Notch Signaling in Adult Kidneys Maintains Segment-Specific Epithelial Cell Types of the Distal Tubules and Collecting Ducts to Ensure Water Homeostasis. J Am Soc Nephrol 2018; 30:110-126. [PMID: 30514723 DOI: 10.1681/asn.2018040440] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 11/07/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Notch signaling is required during kidney development for nephron formation and principal cell fate selection within the collecting ducts. Whether Notch signaling is required in the adult kidney to maintain epithelial diversity, or whether its loss can trigger principal cell transdifferentiation (which could explain acquired diabetes insipidus in patients receiving lithium) is unclear. METHODS To investigate whether loss of Notch signaling can trigger principal cells to lose their identity, we genetically inactivated Notch1 and Notch2, inactivated the Notch signaling target Hes1, or induced expression of a Notch signaling inhibitor in all of the nephron segments and collecting ducts in mice after kidney development. We examined renal function and cell type composition of control littermates and mice with conditional Notch signaling inactivation in adult renal epithelia. In addition, we traced the fate of genetically labeled adult kidney collecting duct principal cells after Hes1 inactivation or lithium treatment. RESULTS Notch signaling was required for maintenance of Aqp2-expressing cells in distal nephron and collecting duct segments in adult kidneys. Fate tracing revealed mature principal cells in the inner stripe of the outer medulla converted to intercalated cells after genetic inactivation of Hes1 and, to a lesser extent, lithium treatment. Hes1 ensured repression of Foxi1 to prevent the intercalated cell program from turning on in mature Aqp2+ cell types. CONCLUSIONS Notch signaling via Hes1 regulates maintenance of mature renal epithelial cell states. Loss of Notch signaling or use of lithium can trigger transdifferentiation of mature principal cells to intercalated cells in adult kidneys.
Collapse
Affiliation(s)
| | | | - Karla Otterpohl
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota
| | - Ishara Ratnayake
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; and
| | - Divya Kota
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; and
| | - Phil Ahrenkiel
- Department of Nanoscience and Nanoengineering, South Dakota School of Mines and Technology, Rapid City, South Dakota; and
| | - Indra Chandrasekar
- Enabling Technologies Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| | - Kameswaran Surendran
- Pediatrics and Rare Diseases Group and .,Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Sioux Falls, South Dakota
| |
Collapse
|
152
|
Jain S, Chen F. Developmental pathology of congenital kidney and urinary tract anomalies. Clin Kidney J 2018; 12:382-399. [PMID: 31198539 PMCID: PMC6543978 DOI: 10.1093/ckj/sfy112] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Indexed: 12/18/2022] Open
Abstract
Congenital anomalies of the kidneys or lower urinary tract (CAKUT) are the most common causes of renal failure in children and account for 25% of end-stage renal disease in adults. The spectrum of anomalies includes renal agenesis; hypoplasia; dysplasia; supernumerary, ectopic or fused kidneys; duplication; ureteropelvic junction obstruction; primary megaureter or ureterovesical junction obstruction; vesicoureteral reflux; ureterocele; and posterior urethral valves. CAKUT originates from developmental defects and can occur in isolation or as part of other syndromes. In recent decades, along with better understanding of the pathological features of the human congenital urinary tract defects, researchers using animal models have provided valuable insights into the pathogenesis of these diseases. However, the genetic causes and etiology of many CAKUT cases remain unknown, presenting challenges in finding effective treatment. Here we provide an overview of the critical steps of normal development of the urinary system, followed by a description of the pathological features of major types of CAKUT with respect to developmental mechanisms of their etiology.
Collapse
Affiliation(s)
- Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| | - Feng Chen
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
| |
Collapse
|
153
|
Stangenberg S, Nguyen LT, Chan YL, Zaky A, Pollock CA, Chen H, Saad S. Maternal L-carnitine supplementation ameliorates renal underdevelopment and epigenetic changes in male mice offspring due to maternal smoking. Clin Exp Pharmacol Physiol 2018; 46:183-193. [PMID: 30290012 DOI: 10.1111/1440-1681.13038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 09/18/2018] [Accepted: 09/27/2018] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Epidemiological and animal studies showed that L-carnitine (LC) supplementation can ameliorate oxidative stress-induced tissues damage. We have previously shown that maternal cigarette smoke exposure (SE) can increase renal oxidative stress in newborn offspring with postnatal kidney underdevelopment and renal dysfunction in adulthood, which were normalised by LC administration in the SE dams during pregnancy. Exposure to an adverse intrauterine environment may lead to alteration in the epigenome, a mechanism by which adverse prenatal conditions increase the susceptibility to chronic disease later in life. The current study aimed to determine whether maternal SE induces epigenetic changes in the offspring's kidney are associated with renal underdevelopment, and the protective effect of maternal LC supplementation. METHOD Female Balb/c mice (7 weeks) were exposed to cigarette smoke (SE) or air (Sham) for 6 weeks prior to mating, during gestation and lactation. A subgroup of the SE dams received LC via drinking water (SE + LC, 1.5 mmol/L) throughout gestation and lactation. Male offspring were studied at postnatal day (P)1, P20, and 13 weeks. RESULTS Maternal SE altered the expression of renal development markers glial cell line-derived neurotrophic factor and fibroblast growth factor 2, which were associated with increased renal global DNA methylation and DNA methyltransferase 1 mRNA expression at birth. These disorders were reversed by maternal LC administration. CONCLUSION The effect of maternal SE on renal underdevelopment involves global epigenetic alterations from birth, which can be prevented by maternal LC supplementation.
Collapse
Affiliation(s)
- Stefanie Stangenberg
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Long The Nguyen
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Yik Lung Chan
- RCMB, Woolcock Institute of Medical Research, The University of Sydney, Sydney, New South Wales, Australia
| | - Amgad Zaky
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Carol A Pollock
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Hui Chen
- Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Sonia Saad
- Renal Group, Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, New South Wales, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
154
|
Phua YL, Clugston A, Chen KH, Kostka D, Ho J. Small non-coding RNA expression in mouse nephrogenic mesenchymal progenitors. Sci Data 2018; 5:180218. [PMID: 30422124 PMCID: PMC6233257 DOI: 10.1038/sdata.2018.218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/15/2018] [Indexed: 01/02/2023] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are essential for the regulation of gene expression and play critical roles in human health and disease. Here we present comprehensive miRNA profiling data for mouse nephrogenic mesenchymal progenitors, a population of cells enriched for nephron progenitors that give rise to most cell-types of the nephron, the functional unit of the kidney. We describe a miRNA expression in nephrogenic mesenchymal progenitors, with 162 miRNAs differentially expressed in progenitors when compared to whole kidney. We also annotated 49 novel miRNAs in the developing kidney and experimentally validated 4 of them. Our data are available as a public resource, so that it can be integrated into future studies and analyzed in the context of other functional and epigenomic data in kidney development. Specifically, it will be useful in the effort to shed light on molecular mechanisms underlying processes essential for normal kidney development, like nephron progenitor specification, self-renewal and differentiation.
Collapse
Affiliation(s)
- Yu Leng Phua
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA
| | - Andrew Clugston
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin Hong Chen
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA
| | - Dennis Kostka
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Developmental Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Department of Computational & Systems Biology and Pittsburgh Center for Evolutionary Biology and Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jacqueline Ho
- Rangos Research Center, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA, USA.,Department of Pediatrics, Division of Nephrology, University of Pittsburgh School of Medicine, PA, USA
| |
Collapse
|
155
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
156
|
Lyu Z, Mao Z, Li Q, Xia Y, Liu Y, He Q, Wang Y, Zhao H, Lu Z, Zhou Q. PPARγ maintains the metabolic heterogeneity and homeostasis of renal tubules. EBioMedicine 2018; 38:178-190. [PMID: 30420298 PMCID: PMC6306377 DOI: 10.1016/j.ebiom.2018.10.072] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 10/22/2018] [Accepted: 10/31/2018] [Indexed: 02/03/2023] Open
Abstract
Background The renal tubules, which have distant metabolic features and functions in different segments, reabsorb >99% of approximately 180 l of water and 25,000 mmol of Na + daily. Defective metabolism in renal tubules is involved in the pathobiology of kidney diseases. However, the mechanisms underlying the metabolic regulation in renal tubules remain to be defined. Methods We quantitatively compared the proteomes of the isolated proximal tubules (PT) and distal tubules (DT) from C57BL/6 mouse using tandem mass tag (TMT) labeling-based quantitative mass spectrometry. Bioinformatics analysis of the differentially expressed proteins revealed the significant differences between PT and DT in metabolism pathway. We also performed in vitro and in vivo assays to investigate the molecular mechanism underlying the distant metabolic features in PT and DT. Findings We demonstrate that the renal proximal tubule (PT) has high expression of lipid metabolism enzymes, which is transcriptionally upregulated by abundantly expressed PPARα/γ. In contrast, the renal distal tubule (DT) has elevated glycolytic enzyme expression, which is mediated by highly expressed c-Myc. Importantly, PPARγ transcriptionally enhances the protease iRhom2 expression in PT, which suppresses EGF expression and secretion and subsequent EGFR-dependent glycolytic gene expression and glycolysis. PPARγ inhibition reduces iRhom2 expression and increases EGF and GLUT1 expression in PT in mice, resulting in renal tubule hypertrophy, tubulointerstitial fibrosis and damaged kidney functions, which are rescued by 2-deoxy-d-glucose treatment. Interpretation These findings delineate instrumental mechanisms underlying the active lipid metabolism and suppressed glycolysis in PT and active glycolysis in DT and reveal critical roles for PPARs and c-Myc in maintaining renal metabolic homeostasis. FUND: This work was supported by the National Natural Science Foundation of China (grants 81572076 and 81873932; to Q.Z.), the Applied Development Program of the Science and Technology Committee of Chongqing (cstc2014yykfB10003; Q.Z.), the Program of Populace Creativities Workshops of the Science and Technology Committee of Chongqing (Q.Z.), the special demonstration programs for innovation and application of techniques (cstc2018jscx-mszdX0022) from the Science and Technology Committee of Chongqing (Q.Z.).
Collapse
Affiliation(s)
- Zhongshi Lyu
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Zhaomin Mao
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China; Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Qianyin Li
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yan Xia
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yamin Liu
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Qingling He
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Hui Zhao
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - Zhimin Lu
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, USA
| | - Qin Zhou
- The Division of Molecular Nephrology, The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China.
| |
Collapse
|
157
|
Bajaj P, Rodrigues AD, Steppan CM, Engle SJ, Mathialagan S, Schroeter T. Human Pluripotent Stem Cell-Derived Kidney Model for Nephrotoxicity Studies. Drug Metab Dispos 2018; 46:1703-1711. [PMID: 30171163 DOI: 10.1124/dmd.118.082727] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/28/2018] [Indexed: 02/13/2025] Open
Abstract
Current in vitro models for identifying nephrotoxins are poorly predictive. We differentiated human pluripotent stem cells (hPSCs) into three-dimensional, multicellular structures containing proximal tubule cells (PTCs) and podocytes and evaluated them as a platform for predicting nephrotoxicity. The PTCs showed megalin-dependent, cubilin-mediated endocytosis of fluorescently labeled dextran and active gamma-glutamyl transpeptidase enzymes. Transporters from both the ATP-binding cassette (ABC) and the solute carrier (SLC) families were present at physiological levels in the differentiated cells, but important renal transporters such as organic anion transporter 1 (OAT1), OAT3, and organic cation transporter 2 (OCT2) were present only at lower levels. Radioactive uptake studies confirmed the functional activity of organic cation transporter, novel, type 2 (OCTN2), organic anion transporter polypeptide 4C1 (OATP4C1), and OCTs/multidrug and toxin extrusion proteins (MATEs). When treated with 10 pharmacologic agents as a test of the platform, the known nephrotoxic compounds were distinguished from the more benign compounds by an increase in tubular (PTC, kidney injury molecule 1 (KIM-1), and heme oxygenase 1 (HO-1)) and glomerular (nephrin [NPHS1]/Wilms tumor protein [WT1]) markers associated with nephrotoxicity, and we were able to distinguish the type of nephrotoxin by examining the relative levels of these markers. Given the functions demonstrated and with improved expression of key renal transporters, this hPSC-derived in vitro kidney model shows promise as a platform for detection of mechanistically different nephrotoxins.
Collapse
Affiliation(s)
- Piyush Bajaj
- Discovery Sciences (P.B., C.M.S., S.J.E., T.S.) and Pharmacokinetics, Dynamics, and Metabolism (A.D.R., S.M.), Pfizer Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - A David Rodrigues
- Discovery Sciences (P.B., C.M.S., S.J.E., T.S.) and Pharmacokinetics, Dynamics, and Metabolism (A.D.R., S.M.), Pfizer Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Claire M Steppan
- Discovery Sciences (P.B., C.M.S., S.J.E., T.S.) and Pharmacokinetics, Dynamics, and Metabolism (A.D.R., S.M.), Pfizer Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sandra J Engle
- Discovery Sciences (P.B., C.M.S., S.J.E., T.S.) and Pharmacokinetics, Dynamics, and Metabolism (A.D.R., S.M.), Pfizer Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Discovery Sciences (P.B., C.M.S., S.J.E., T.S.) and Pharmacokinetics, Dynamics, and Metabolism (A.D.R., S.M.), Pfizer Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Thomas Schroeter
- Discovery Sciences (P.B., C.M.S., S.J.E., T.S.) and Pharmacokinetics, Dynamics, and Metabolism (A.D.R., S.M.), Pfizer Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
158
|
Ide S, Finer G, Maezawa Y, Onay T, Souma T, Scott R, Ide K, Akimoto Y, Li C, Ye M, Zhao X, Baba Y, Minamizuka T, Jin J, Takemoto M, Yokote K, Quaggin SE. Transcription Factor 21 Is Required for Branching Morphogenesis and Regulates the Gdnf-Axis in Kidney Development. J Am Soc Nephrol 2018; 29:2795-2808. [PMID: 30377232 DOI: 10.1681/asn.2017121278] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 09/27/2018] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND The mammalian kidney develops through reciprocal inductive signals between the metanephric mesenchyme and ureteric bud. Transcription factor 21 (Tcf21) is highly expressed in the metanephric mesenchyme, including Six2-expressing cap mesenchyme and Foxd1-expressing stromal mesenchyme. Tcf21 knockout mice die in the perinatal period from severe renal hypodysplasia. In humans, Tcf21 mRNA levels are reduced in renal tissue from human fetuses with renal dysplasia. The molecular mechanisms underlying these renal defects are not yet known. METHODS Using a variety of techniques to assess kidney development and gene expression, we compared the phenotypes of wild-type mice, mice with germline deletion of the Tcf21 gene, mice with stromal mesenchyme-specific Tcf21 deletion, and mice with cap mesenchyme-specific Tcf21 deletion. RESULTS Germline deletion of Tcf21 leads to impaired ureteric bud branching and is accompanied by downregulated expression of Gdnf-Ret-Wnt11, a key pathway required for branching morphogenesis. Selective removal of Tcf21 from the renal stroma is also associated with attenuation of the Gdnf signaling axis and leads to a defect in ureteric bud branching, a paucity of collecting ducts, and a defect in urine concentration capacity. In contrast, deletion of Tcf21 from the cap mesenchyme leads to abnormal glomerulogenesis and massive proteinuria, but no downregulation of Gdnf-Ret-Wnt11 or obvious defect in branching. CONCLUSIONS Our findings indicate that Tcf21 has distinct roles in the cap mesenchyme and stromal mesenchyme compartments during kidney development and suggest that Tcf21 regulates key molecular pathways required for branching morphogenesis.
Collapse
Affiliation(s)
- Shintaro Ide
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Gal Finer
- Division of Kidney Diseases, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Feinberg Cardiovascular and Renal Research Institute and
| | - Yoshiro Maezawa
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan;
| | - Tuncer Onay
- Feinberg Cardiovascular and Renal Research Institute and.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Tomokazu Souma
- Feinberg Cardiovascular and Renal Research Institute and.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Rizaldy Scott
- Feinberg Cardiovascular and Renal Research Institute and.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Kana Ide
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Yoshihiro Akimoto
- Department of Anatomy, Kyorin University School of Medicine, Tokyo, Japan
| | - Chengjin Li
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada; and
| | - Minghao Ye
- Feinberg Cardiovascular and Renal Research Institute and.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Xiangmin Zhao
- Division of Kidney Diseases, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois.,Feinberg Cardiovascular and Renal Research Institute and
| | - Yusuke Baba
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan;
| | - Takuya Minamizuka
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan;
| | - Jing Jin
- Feinberg Cardiovascular and Renal Research Institute and.,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Minoru Takemoto
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan.,Division of Diabetes, Metabolism and Endocrinology, International University of Health and Welfare, Narita, Japan
| | - Koutaro Yokote
- Department of Clinical Cell Biology and Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Susan E Quaggin
- Feinberg Cardiovascular and Renal Research Institute and .,Division of Nephrology/Hypertension, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
159
|
He Q, Chen L, Liu Y, Wu Y, Ni D, Liu J, Hu Y, Gu Y, Xie Y, Zhou Q, Li Q. Gulo regulates the proliferation, apoptosis and mesenchymal-to-epithelial transformation of metanephric mesenchyme cells via inhibiting Six2. Biochem Biophys Res Commun 2018; 504:885-891. [PMID: 30219227 DOI: 10.1016/j.bbrc.2018.08.062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022]
Abstract
During kidney development, the balance between self-renewal and differentiation of metanephric mesenchyme (MM) cells, mainly regulated by Sine oculis-related homeobox 2 (Six2), is critical for forming mature kidney. L-gulono-γ-lactone oxidase (Gulo), a crucial enzyme for vitamin C synthesis, reveals a different expression at various stages during kidney development, but its function in the early renal development remains unknown. In this work, we aim to study the role of Gulo in MM cells at two differentiation stages. We found that Gulo expression in undifferentiated MM (mK3) cells was lower than in differentiated MM (mK4) cells. Over-expression of Gulo can promote mesenchymal-to-epithelial transformation (MET) and apoptosis and inhibit the proliferation in mK3 cells. Knock-down of Gulo in mK4 cells made its epithelial character cells unstabilized, facilitated the proliferation and restrained the apoptosis. Furthermore, we found that Six2 was negatively regulated by Gulo, and over-expression or knock-down of Six2 was able to rescue partially the MET, proliferation and apoptosis of MM cells caused by Gulo. In conclusion, these findings reveal that Gulo promotes the MET and apoptosis, and inhibits proliferation in MM cells by down-regulating Six2.
Collapse
Affiliation(s)
- Qingling He
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Lei Chen
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yamin Liu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yafei Wu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Dongsheng Ni
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Jianing Liu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yanxia Hu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yuping Gu
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Yajun Xie
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Qin Zhou
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| | - Qianyin Li
- The M.O.E. Key Laboratory of Laboratory Medical Diagnostics, The College of Laboratory Medicine, Chongqing Medical University, #1 Yixueyuan Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
160
|
Tajiri S, Yamanaka S, Fujimoto T, Matsumoto K, Taguchi A, Nishinakamura R, Okano HJ, Yokoo T. Regenerative potential of induced pluripotent stem cells derived from patients undergoing haemodialysis in kidney regeneration. Sci Rep 2018; 8:14919. [PMID: 30297790 PMCID: PMC6175865 DOI: 10.1038/s41598-018-33256-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 09/26/2018] [Indexed: 12/11/2022] Open
Abstract
Kidney regeneration from pluripotent stem cells is receiving a lot of attention because limited treatments are currently available for chronic kidney disease (CKD). It has been shown that uremic state in CKD is toxic to somatic stem/progenitor cells, such as endothelial progenitor and mesenchymal stem cells, affecting their differentiation and angiogenic potential. Recent studies reported that specific abnormalities caused by the non-inherited disease are often retained in induced pluripotent stem cell (iPSC)-derived products obtained from patients. Thus, it is indispensable to first assess whether iPSCs derived from patients with CKD due to non-inherited disease (CKD-iPSCs) have the ability to generate kidneys. In this study, we generated iPSCs from patients undergoing haemodialysis due to diabetes nephropathy and glomerulonephritis (HD-iPSCs) as representatives of CKD-iPSCs or from healthy controls (HC-iPSCs). HD-iPSCs differentiated into nephron progenitor cells (NPCs) with similar efficiency to HC-iPSCs. Additionally, HD-iPSC-derived NPCs expressed comparable levels of NPC markers and differentiated into vascularised glomeruli upon transplantation into mice, as HC-iPSC-derived NPCs. Our results indicate the potential of HD-iPSCs as a feasible cell source for kidney regeneration. This is the first study paving the way for CKD patient-stem cell-derived kidney regeneration, emphasising the potential of CKD-iPSCs.
Collapse
Affiliation(s)
- Susumu Tajiri
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Shuichiro Yamanaka
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Toshinari Fujimoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Kei Matsumoto
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Atsuhiro Taguchi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1, Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Ihnestraße 63-73, 14195, Berlin, Germany
| | - Ryuichi Nishinakamura
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, 2-2-1, Honjo, Chuo-ku, Kumamoto, 860-0811, Japan
| | - Hirotaka James Okano
- Division of Regenerative Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan
| | - Takashi Yokoo
- Division of Nephrology and Hypertension, Department of Internal Medicine, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-ku, Tokyo, 105-8461, Japan.
| |
Collapse
|
161
|
Recapitulating kidney development: Progress and challenges. Semin Cell Dev Biol 2018; 91:153-168. [PMID: 30184476 DOI: 10.1016/j.semcdb.2018.08.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 12/14/2022]
Abstract
Decades of research into the molecular and cellular regulation of kidney morphogenesis in rodent models, particularly the mouse, has provided both an atlas of the mammalian kidney and a roadmap for recreating kidney cell types with potential applications for the treatment of kidney disease. With advances in both our capacity to maintain nephron progenitors in culture, reprogram to kidney cell types and direct the differentiation of human pluripotent stem cells to kidney endpoints, renal regeneration via cellular therapy or tissue engineering may be possible. Human kidney models also have potential for disease modelling and drug screening. Such applications will rely upon the accuracy of the model at the cellular level and the capacity for stem-cell derived kidney tissue to recapitulate both normal and diseased kidney tissue. In this review, we will discuss the available cell sources, how well they model the human kidney and how far we are from application either as models or for tissue engineering.
Collapse
|
162
|
Nishikawa M, Yuri S, Kimura H, Yanagawa N, Hamon M, Hauser P, Zhao L, Jo OD, Yanagawa N. Comprehensive analysis of chromatin signature and transcriptome uncovers functional lncRNAs expressed in nephron progenitor cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1862:58-70. [PMID: 30416088 DOI: 10.1016/j.bbagrm.2018.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 08/25/2018] [Accepted: 09/12/2018] [Indexed: 01/21/2023]
Abstract
Emerging evidence from recent studies has unraveled the roles of long noncoding RNAs (lncRNAs) in the function of various tissues. However, little is known about the roles of lncRNAs in kidney development. In our present study, we aimed to identify functional lncRNAs in one of the three lineages of kidney progenitor cells, i.e., metanephric mesenchymal (MM) cells. We conducted comprehensive analyses of the chromatin signature and transcriptome by RNA-seq and ChIP-seq. We found seventeen lncRNAs that were expressed specifically in MM cells with an active chromatin signature, while remaining silenced in a bivalent chromatin state in non-MM cells. Out of these MM specific lncRNAs, we identified a lncRNA, Gm29418, in a distal enhancer region of Six2, a key regulatory gene of MM cells. We further identified three transcript variants of Gm29418 by Rapid Amplification of cDNA Ends (RACE), and confirmed that the transcription-start-sites (TSSs) of these variants were consistent with the result of Cap Analysis Gene Expression (CAGE). In support of the enhancer-like function of Gm29418 on Six2 expression, we found that knock-down of Gm29418 by two independent anti-sense locked nucleic acid (LNA) phosphorothioate gapmers suppressed Six2 mRNA expression levels in MM cells. We also found that over-expression of Gm29418 led to an increase in Six2 mRNA expression levels in a mouse MM cell line. In conclusion, we identified a lncRNA, Gm29418, in nephron progenitor cells that has an enhancer-like function on a key regulatory gene, Six2.
Collapse
Affiliation(s)
- Masaki Nishikawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| | - Shunsuke Yuri
- Nara Institute of Science & Technology, Nara 630-0192, Japan
| | | | - Naomi Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Morgan Hamon
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Peter Hauser
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Lifu Zhao
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA
| | - Oak D Jo
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Norimoto Yanagawa
- Medical and Research Services, Greater Los Angeles Veterans Affairs Healthcare System at Sepulveda, North Hills, CA, USA; University of California at Los Angeles, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
163
|
El-Dahr SS, Saifudeen Z. Epigenetic regulation of renal development. Semin Cell Dev Biol 2018; 91:111-118. [PMID: 30172047 DOI: 10.1016/j.semcdb.2018.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 07/19/2018] [Accepted: 08/28/2018] [Indexed: 01/24/2023]
Abstract
Developmental changes in cell fate are tightly regulated by cell-type specific transcription factors. Chromatin reorganization during organismal development ensures dynamic access of developmental regulators to their cognate DNA sequences. Thus, understanding the epigenomic states of promoters and enhancers is of key importance. Recent years have witnessed significant advances in our knowledge of the transcriptional mechanisms of kidney development. Emerging evidence suggests that histone deacetylation by class I HDACs and H3 methylation on lysines 4, 27 and 79 play important roles in regulation of early and late gene expression in the developing kidney. Equally exciting is the realization that nephrogenesis genes in mesenchymal nephron progenitors harbor bivalent chromatin domains which resolve upon differentiation implicating chromatin bivalency in developmental control of gene expression. Here, we review current knowledge of the epigenomic states of nephric cells and current techniques used to study the dynamic chromatin states. These technological advances will provide an unprecedented view of the enhancer landscape during cell fate commitment and help in defining the complex transcriptional networks governing kidney development and disease.
Collapse
Affiliation(s)
- Samir S El-Dahr
- Tulane University School of Medicine, 1430 Tulane Avenue, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA, 70112, USA.
| | - Zubaida Saifudeen
- Tulane University School of Medicine, 1430 Tulane Avenue, Department of Pediatrics, Section of Pediatric Nephrology, New Orleans, LA, 70112, USA.
| |
Collapse
|
164
|
Sivakumar A, Kurpios NA. Transcriptional regulation of cell shape during organ morphogenesis. J Cell Biol 2018; 217:2987-3005. [PMID: 30061107 PMCID: PMC6122985 DOI: 10.1083/jcb.201612115] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 06/11/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023] Open
Abstract
The emerging field of transcriptional regulation of cell shape changes aims to address the critical question of how gene expression programs produce a change in cell shape. Together with cell growth, division, and death, changes in cell shape are essential for organ morphogenesis. Whereas most studies of cell shape focus on posttranslational events involved in protein organization and distribution, cell shape changes can be genetically programmed. This review highlights the essential role of transcriptional regulation of cell shape during morphogenesis of the heart, lungs, gastrointestinal tract, and kidneys. We emphasize the evolutionary conservation of these processes across different model organisms and discuss perspectives on open questions and research avenues that may provide mechanistic insights toward understanding birth defects.
Collapse
Affiliation(s)
- Aravind Sivakumar
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY
| |
Collapse
|
165
|
O'Brien LL. Nephron progenitor cell commitment: Striking the right balance. Semin Cell Dev Biol 2018; 91:94-103. [PMID: 30030141 DOI: 10.1016/j.semcdb.2018.07.017] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 06/29/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
The filtering component of the kidney, the nephron, arises from a single progenitor population. These nephron progenitor cells (NPCs) both self-renew and differentiate throughout the course of kidney development ensuring sufficient nephron endowment. An appropriate balance of these processes must be struck as deficiencies in nephron numbers are associated with hypertension and kidney disease. This review will discuss the mechanisms and molecules supporting NPC maintenance and differentiation. A focus on recent work will highlight new molecular insights into NPC regulation and their dynamic behavior in both space and time.
Collapse
Affiliation(s)
- Lori L O'Brien
- Department of Cell Biology and Physiology, UNC Kidney Center, University of North Carolina at Chapel Hill, 111 Mason Farm Road, Chapel Hill, NC, 27599, United States.
| |
Collapse
|
166
|
Held M, Santeramo I, Wilm B, Murray P, Lévy R. Ex vivo live cell tracking in kidney organoids using light sheet fluorescence microscopy. PLoS One 2018; 13:e0199918. [PMID: 30048451 PMCID: PMC6062017 DOI: 10.1371/journal.pone.0199918] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/16/2018] [Indexed: 12/28/2022] Open
Abstract
Screening cells for their differentiation potential requires a combination of tissue culture models and imaging methods that allow for long-term tracking of the location and function of cells. Embryonic kidney re-aggregation in vitro assays have been established which allow for the monitoring of organotypic cell behaviour in re-aggregated and chimeric renal organoids. However, evaluation of cell integration is hampered by the high photonic load of standard fluorescence microscopy which poses challenges for imaging three-dimensional systems in real-time over a time course. Therefore, we employed light sheet microscopy, a technique that vastly reduces photobleaching and phototoxic effects. We have also developed a new method for culturing the re-aggregates which involves immersed culture, generating organoids which more closely reflect development in vivo. To facilitate imaging from various angles, we embedded the organoids in a freely rotatable hydrogel cylinder. Endpoint fixing and staining were performed to provide additional biomolecular information. We succeeded in imaging labelled cells within re-aggregated kidney organoids over 15 hours and tracking their fate while simultaneously monitoring the development of organotypic morphological structures. Our results show that Wt1-expressing embryonic kidney cells obtained from transgenic mice could integrate into re-aggregated chimeric kidney organoids and contribute to developing nephrons. Furthermore, the nascent proximal tubules that formed in the re-aggregated tissues using the new culture method displayed secretory function, as evidenced by their ability to secrete an organic anion mimic into the tubular lumen.
Collapse
Affiliation(s)
- Marie Held
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Ilaria Santeramo
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Bettina Wilm
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Patricia Murray
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Raphaël Lévy
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
167
|
Geometry of Gene Expression Space of Wilms' Tumors From Human Patients. Neoplasia 2018; 20:871-881. [PMID: 30029183 PMCID: PMC6076422 DOI: 10.1016/j.neo.2018.06.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 06/12/2018] [Accepted: 06/19/2018] [Indexed: 02/05/2023] Open
Abstract
Wilms' tumor is a pediatric malignancy that is thought to originate from faulty kidney development during the embryonic stage. However, there is a large variation between tumors from different patients in both histology and gene expression that is not well characterized. Here we use a meta-analysis of published microarray datasets to show that Favorable Histology Wilms' Tumors (FHWT's) fill a triangle-shaped continuum in gene expression space of which the vertices represent three idealized “archetypes”. We show that these archetypes have predominantly renal blastemal, stromal, and epithelial characteristics and that they correlate well with the three major lineages of the developing embryonic kidney. Moreover, we show that advanced stage tumors shift towards the renal blastemal archetype. These results illustrate the potential of this methodology for characterizing the cellular composition of Wilms' tumors and for assessing disease progression.
Collapse
|
168
|
Becherucci F, Mazzinghi B, Allinovi M, Angelotti ML, Romagnani P. Regenerating the kidney using human pluripotent stem cells and renal progenitors. Expert Opin Biol Ther 2018; 18:795-806. [PMID: 29939787 DOI: 10.1080/14712598.2018.1492546] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Chronic kidney disease is a major health-care problem worldwide and its cost is becoming no longer affordable. Indeed, restoring damaged renal structures or building a new kidney represents an ambitious and ideal alternative to renal replacement therapy. Streams of research have explored the possible application of pluripotent stem cells (SCs) (embryonic SCs and induced pluripotent SCs) in different strategies aimed at regenerate functioning nephrons and at understanding the mechanisms of kidney regeneration. AREAS COVERED In this review, we will focus on the main potential applications of human pluripotent SCs to kidney regeneration, including those leading to rebuilding new kidneys or part of them (organoids, scaffolds, biological microdevices) as well as those aimed at understanding the pathophysiological mechanisms of renal disease and regenerative processes (modeling of kidney disease, genome editing). Moreover, we will discuss the role of endogenous renal progenitors cells in order to understand and promote kidney regeneration, as an attractive alternative to pluripotent SCs. EXPERT OPINION Opportunities and pitfalls of all these strategies will be underlined, finally leading to the conclusion that a deeper knowledge of the biology of pluripotent SCs is mandatory, in order to allow us to hypothesize their clinical application.
Collapse
Affiliation(s)
- Francesca Becherucci
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy
| | - Benedetta Mazzinghi
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy
| | - Marco Allinovi
- b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Maria Lucia Angelotti
- b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Paola Romagnani
- a Nephrology and Dialysis Unit , Meyer Children's University Hospital , Florence , Italy.,b Department of Biomedical Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| |
Collapse
|
169
|
An optimal serum-free defined condition for in vitro culture of kidney organoids. Biochem Biophys Res Commun 2018; 501:996-1002. [DOI: 10.1016/j.bbrc.2018.05.098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/15/2018] [Indexed: 12/21/2022]
|
170
|
Boivin FJ, Bridgewater D. β-Catenin in stromal progenitors controls medullary stromal development. Am J Physiol Renal Physiol 2018; 314:F1177-F1187. [DOI: 10.1152/ajprenal.00282.2017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The renal stroma is a population of matrix-producing fibroblast cells that serves as a structural framework for the kidney parenchyma. The stroma also regulates branching morphogenesis and nephrogenesis. In the mature kidney, the stroma forms at least three distinct cell populations: the capsular, cortical, and medullary stroma. These distinct stromal populations have important functions in kidney development, maintenance of kidney function, and disease progression. However, the development, differentiation, and maintenance of the distinct stroma populations are not well defined. Using a mouse model with β-catenin deficiency in the stroma cell population, we demonstrate that β-catenin is not involved in the formation of the stromal progenitors nor in the formation of the cortical stroma population. In contrast, β-catenin does control the differentiation of stromal progenitors to form the medullary stroma. In the absence of stromal β-catenin, there is a marked reduction of medullary stromal markers. As kidney development continues, the maldifferentiated stromal cells locate deeper within the kidney tissue and are eliminated by the activation of an intrinsic apoptotic program. This leads to significant reductions in the medullary stroma population and the lack of medulla formation. Taken together, our results indicate that stromal β-catenin is essential for kidney development by regulating medulla formation through the differentiation of medullary stromal cells.
Collapse
Affiliation(s)
- Felix J. Boivin
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Darren Bridgewater
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
171
|
Lambert B, MacLean AL, Fletcher AG, Combes AN, Little MH, Byrne HM. Bayesian inference of agent-based models: a tool for studying kidney branching morphogenesis. J Math Biol 2018; 76:1673-1697. [PMID: 29392399 PMCID: PMC5906521 DOI: 10.1007/s00285-018-1208-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 01/02/2018] [Indexed: 12/11/2022]
Abstract
The adult mammalian kidney has a complex, highly-branched collecting duct epithelium that arises as a ureteric bud sidebranch from an epithelial tube known as the nephric duct. Subsequent branching of the ureteric bud to form the collecting duct tree is regulated by subcellular interactions between the epithelium and a population of mesenchymal cells that surround the tips of outgrowing branches. The mesenchymal cells produce glial cell-line derived neurotrophic factor (GDNF), that binds with RET receptors on the surface of the epithelial cells to stimulate several subcellular pathways in the epithelium. Such interactions are known to be a prerequisite for normal branching development, although competing theories exist for their role in morphogenesis. Here we introduce the first agent-based model of ex vivo kidney uretic branching. Through comparison with experimental data, we show that growth factor-regulated growth mechanisms can explain early epithelial cell branching, but only if epithelial cell division depends in a switch-like way on the local growth factor concentration; cell division occurring only if the driving growth factor level exceeds a threshold. We also show how a recently-developed method, "Approximate Approximate Bayesian Computation", can be used to infer key model parameters, and reveal the dependency between the parameters controlling a growth factor-dependent growth switch. These results are consistent with a requirement for signals controlling proliferation and chemotaxis, both of which are previously identified roles for GDNF.
Collapse
Affiliation(s)
- Ben Lambert
- Department of Zoology, University of Oxford, Oxford, UK.
| | - Adam L MacLean
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, UK
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
| | - Alexander G Fletcher
- School of Mathematics and Statistics, University of Sheffield, Hicks Building, Hounsfield Road, Sheffield, S3 7RH, UK
- Bateson Centre, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Alexander N Combes
- Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, VIC, 3010, Australia
- Murdoch Childrens Research Institute, Flemington Rd, Parkville, Melbourne, VIC, 3052, Australia
| | - Melissa H Little
- Murdoch Childrens Research Institute, Flemington Rd, Parkville, Melbourne, VIC, 3052, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Helen M Byrne
- Mathematical Institute, University of Oxford, Andrew Wiles Building, Woodstock Road, Oxford, UK
| |
Collapse
|
172
|
Rahman MS, Spitzhorn LS, Wruck W, Hagenbeck C, Balan P, Graffmann N, Bohndorf M, Ncube A, Guillot PV, Fehm T, Adjaye J. The presence of human mesenchymal stem cells of renal origin in amniotic fluid increases with gestational time. Stem Cell Res Ther 2018; 9:113. [PMID: 29695308 PMCID: PMC5918774 DOI: 10.1186/s13287-018-0864-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 03/19/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022] Open
Abstract
Background Established therapies for managing kidney dysfunction such as kidney dialysis and transplantation are limited due to the shortage of compatible donated organs and high costs. Stem cell-based therapies are currently under investigation as an alternative treatment option. As amniotic fluid is composed of fetal urine harboring mesenchymal stem cells (AF-MSCs), we hypothesized that third-trimester amniotic fluid could be a novel source of renal progenitor and differentiated cells. Methods Human third-trimester amniotic fluid cells (AFCs) were isolated and cultured in distinct media. These cells were characterized as renal progenitor cells with respect to cell morphology, cell surface marker expression, transcriptome and differentiation into chondrocytes, osteoblasts and adipocytes. To test for renal function, a comparative albumin endocytosis assay was performed using AF-MSCs and commercially available renal cells derived from kidney biopsies. Comparative transcriptome analyses of first, second and third trimester-derived AF-MSCs were conducted to monitor expression of renal-related genes. Results Regardless of the media used, AFCs showed expression of pluripotency-associated markers such as SSEA4, TRA-1-60, TRA-1-81 and C-Kit. They also express the mesenchymal marker Vimentin. Immunophenotyping confirmed that third-trimester AFCs are bona fide MSCs. AF-MSCs expressed the master renal progenitor markers SIX2 and CITED1, in addition to typical renal proteins such as PODXL, LHX1, BRN1 and PAX8. Albumin endocytosis assays demonstrated the functionality of AF-MSCs as renal cells. Additionally, upregulated expression of BMP7 and downregulation of WT1, CD133, SIX2 and C-Kit were observed upon activation of WNT signaling by treatment with the GSK-3 inhibitor CHIR99201. Transcriptome analysis and semiquantitative PCR revealed increasing expression levels of renal-specific genes (e.g., SALL1, HNF4B, SIX2) with gestational time. Moreover, AF-MSCs shared more genes with human kidney cells than with native MSCs and gene ontology terms revealed involvement of biological processes associated with kidney morphogenesis. Conclusions Third-trimester amniotic fluid contains AF-MSCs of renal origin and this novel source of kidney progenitors may have enormous future potentials for disease modeling, renal repair and drug screening. Electronic supplementary material The online version of this article (10.1186/s13287-018-0864-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Md Shaifur Rahman
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Lucas-Sebastian Spitzhorn
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Carsten Hagenbeck
- Department of Obstetrics and Gynaecology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Percy Balan
- Department of Obstetrics and Gynaecology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Nina Graffmann
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Martina Bohndorf
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Audrey Ncube
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Pascale V Guillot
- Institute for Women's Health, Maternal and Fetal Medicine Department, University College London, London, WC1E 6HX, UK
| | - Tanja Fehm
- Department of Obstetrics and Gynaecology, Medical Faculty, Heinrich Heine University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich Heine University, Moorenstraße 5, 40225, Düsseldorf, Germany.
| |
Collapse
|
173
|
Hussein KH, Saleh T, Ahmed E, Kwak HH, Park KM, Yang SR, Kang BJ, Choi KY, Kang KS, Woo HM. Biocompatibility and hemocompatibility of efficiently decellularized whole porcine kidney for tissue engineering. J Biomed Mater Res A 2018; 106:2034-2047. [PMID: 29569325 DOI: 10.1002/jbm.a.36407] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 02/15/2018] [Accepted: 03/15/2018] [Indexed: 12/14/2022]
Abstract
Whole kidney decellularization is a promising approach in regenerative medicine for engineering a functional organ. The reaction of the potential host depends on the biocompatibility of these decellularized constructs. Despite the proven ability of decellularized kidney scaffolds to guide cell attachment and growth, little is known about biocompatibility and hemocompatibility of these scaffolds. Our aim is to prepare decellularized kidneys of a clinically relevant size and evaluate its biocompatibility and hemocompatibility. Porcine kidneys were cannulated via the renal artery, and then perfused with 0.1% sodium dodecyl sulfate solution. Hematoxylin and eosin as well as DAPI staining confirmed cellular clearance from native kidneys in addition to preservation of the microstructure. SEM confirmed the absence of any cellular content within the scaffold, which is maintained in a well-organized 3D architecture. Decellularized kidneys retained the intact renal vasculature upon examination with contrast radiography. The essential structural extracellular matrix molecules were well-preserved. Scaffolds were susceptible to enzymatic degradation upon collagenase treatment. Scaffolds showed a good hemocompatibility when exposed to porcine blood. Decellularization was efficient to remove 97.7% of DNA from native kidneys in addition to the immunogenic and pathogenic antigens. Scaffolds did not induce the human immune response in vitro. Decellularized kidneys were non-cytotoxic to pig kidney cells (PKs). PKs were able to grow and proliferate within the decellularized renal scaffolds with maintaining a higher function than cells grown as monolayers. Thus, we have developed a rapid decellularization technique for generating biocompatible kidney scaffolds that represents a step toward development of a transplantable organ. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2034-2047, 2018.
Collapse
Affiliation(s)
- Kamal Hany Hussein
- Department of Animal Surgery, College of Veterinary Medicine, Assiut University, Assiut, 71515, Egypt
| | - Tarek Saleh
- Stem Cell Institute, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea.,Department of Surgery, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| | - Ebtehal Ahmed
- Stem Cell Institute, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea.,Department of Surgery, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| | - Ho-Hyun Kwak
- Stem Cell Institute, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea.,Department of Surgery, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| | - Kyung-Mee Park
- Department of Surgery, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Se-Ran Yang
- Stem Cell Institute, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea.,Department of Thoracic and Cardiovascular Surgery, College of Medicine, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| | - Byung-Jae Kang
- Department of Surgery, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| | - Ki-Young Choi
- Department of Controlled Agriculture, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| | - Kyung-Sun Kang
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.,Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Heung-Myong Woo
- Stem Cell Institute, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea.,Department of Surgery, College of Veterinary Medicine, Kangwon National University, Chuncheon, Gangwon, 200-701, Republic of Korea
| |
Collapse
|
174
|
Abstract
PURPOSE OF REVIEW Podocytes are critical components of the nephron filtration barrier and are depleted in many kidney injuries and disease states. Terminally differentiated adult podocytes are highly specialized, postmitotic cells, raising the question of whether the body has any ability to regenerate lost podocytes. This timely question has recently been illuminated by a series of innovative studies. Here, we review recent progress on this topic of significant interest and debate. RECENT FINDINGS The innovation of genetic labeling techniques enables fate tracing of individual podocytes, providing the strongest evidence yet that podocytes can be replaced by nearby progenitor cells. In particular, two progenitor pools have recently been identified in multiple studies: parietal epithelial cells and cells of renin lineage. These studies furthermore suggest that podocyte regeneration can be enhanced using ex-vivo or pharmacological interventions. SUMMARY Recent studies indicate that the podocyte compartment is more dynamic than previously believed. Bidirectional exchange with neighboring cellular compartments provides a mechanism for podocyte replacement. Based on these findings, we propose a set of criteria for evaluating podocyte regeneration and suggest that restoration of podocyte number to a subsclerotic threshold be targeted as a potentially achievable clinical goal.
Collapse
|
175
|
Desai PB, San Agustin JT, Stuck MW, Jonassen JA, Bates CM, Pazour GJ. Ift25 is not a cystic kidney disease gene but is required for early steps of kidney development. Mech Dev 2018; 151:10-17. [PMID: 29626631 DOI: 10.1016/j.mod.2018.04.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 03/13/2018] [Accepted: 04/01/2018] [Indexed: 12/26/2022]
Abstract
Eukaryotic cilia are assembled by intraflagellar transport (IFT) where large protein complexes called IFT particles move ciliary components from the cell body to the cilium. Defects in most IFT particle proteins disrupt ciliary assembly and cause mid gestational lethality in the mouse. IFT25 and IFT27 are unusual components of IFT-B in that they are not required for ciliary assembly and mutant mice survive to term. The mutants die shortly after birth with numerous organ defects including duplex kidneys. Completely duplex kidneys result from defects in ureteric bud formation at the earliest steps of metanephric kidney development. Ureteric bud initiation is a highly regulated process involving reciprocal signaling between the ureteric epithelium and the overlying metanephric mesenchyme with regulation by the peri-Wolffian duct stroma. The finding of duplex kidney in Ift25 and Ift27 mutants suggests functions for these genes in regulation of ureteric bud initiation. Typically the deletion of IFT genes in the kidney causes rapid cyst growth in the early postnatal period. In contrast, the loss of Ift25 results in smaller kidneys, which show only mild tubule dilations that become apparent in adulthood. The smaller kidneys appear to result from reduced branching in the developing metanephric kidney. This work indicates that IFT25 and IFT27 are important players in the early development of the kidney and suggest that duplex kidney is part of the ciliopathy spectrum.
Collapse
Affiliation(s)
- Paurav B Desai
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Suite 213, 373 Plantation Street, Worcester, MA 01605, United States
| | - Jovenal T San Agustin
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Suite 213, 373 Plantation Street, Worcester, MA 01605, United States
| | - Michael W Stuck
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Suite 213, 373 Plantation Street, Worcester, MA 01605, United States
| | - Julie A Jonassen
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655, United States
| | - Carlton M Bates
- Department of Pediatrics, University of Pittsburgh, Children's Hospital of Pittsburgh of UPMC, 5130 Rangos Research Center, 4401 Penn Avenue, Pittsburgh, PA 15224, United States
| | - Gregory J Pazour
- Program in Molecular Medicine, University of Massachusetts Medical School, Biotech II, Suite 213, 373 Plantation Street, Worcester, MA 01605, United States.
| |
Collapse
|
176
|
Lindström NO, McMahon JA, Guo J, Tran T, Guo Q, Rutledge E, Parvez RK, Saribekyan G, Schuler RE, Liao C, Kim AD, Abdelhalim A, Ruffins SW, Thornton ME, Baskin L, Grubbs B, Kesselman C, McMahon AP. Conserved and Divergent Features of Human and Mouse Kidney Organogenesis. J Am Soc Nephrol 2018; 29:785-805. [PMID: 29449453 DOI: 10.1681/asn.2017080887] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 11/27/2017] [Indexed: 01/22/2023] Open
Abstract
Human kidney function is underpinned by approximately 1,000,000 nephrons, although the number varies substantially, and low nephron number is linked to disease. Human kidney development initiates around 4 weeks of gestation and ends around 34-37 weeks of gestation. Over this period, a reiterative inductive process establishes the nephron complement. Studies have provided insightful anatomic descriptions of human kidney development, but the limited histologic views are not readily accessible to a broad audience. In this first paper in a series providing comprehensive insight into human kidney formation, we examined human kidney development in 135 anonymously donated human kidney specimens. We documented kidney development at a macroscopic and cellular level through histologic analysis, RNA in situ hybridization, immunofluorescence studies, and transcriptional profiling, contrasting human development (4-23 weeks) with mouse development at selected stages (embryonic day 15.5 and postnatal day 2). The high-resolution histologic interactive atlas of human kidney organogenesis generated can be viewed at the GUDMAP database (www.gudmap.org) together with three-dimensional reconstructions of key components of the data herein. At the anatomic level, human and mouse kidney development differ in timing, scale, and global features such as lobe formation and progenitor niche organization. The data also highlight differences in molecular and cellular features, including the expression and cellular distribution of anchor gene markers used to identify key cell types in mouse kidney studies. These data will facilitate and inform in vitro efforts to generate human kidney structures and comparative functional analyses across mammalian species.
Collapse
Affiliation(s)
- Nils O Lindström
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Jill A McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Jinjin Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Tracy Tran
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Qiuyu Guo
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Elisabeth Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Riana K Parvez
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Gohar Saribekyan
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | | | - Christopher Liao
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Albert D Kim
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Ahmed Abdelhalim
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Seth W Ruffins
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine
| | - Matthew E Thornton
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, California; and
| | - Laurence Baskin
- Department of Urology and Pediatrics, University of California San Francisco, San Francisco, California
| | - Brendan Grubbs
- Maternal Fetal Medicine Division, University of Southern California, Los Angeles, California; and
| | - Carl Kesselman
- Information Sciences Institute, Viterbi School of Engineering.,Epstein Department of Industrial and Systems Engineering and Information Sciences Institute, Viterbi School of Engineering and Department of Preventive Medicine, Keck School of Medicine, and
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine,
| |
Collapse
|
177
|
Hsieh WC, Ramadesikan S, Fekete D, Aguilar RC. Kidney-differentiated cells derived from Lowe Syndrome patient's iPSCs show ciliogenesis defects and Six2 retention at the Golgi complex. PLoS One 2018; 13:e0192635. [PMID: 29444177 PMCID: PMC5812626 DOI: 10.1371/journal.pone.0192635] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 01/26/2018] [Indexed: 12/11/2022] Open
Abstract
Lowe syndrome is an X-linked condition characterized by congenital cataracts, neurological abnormalities and kidney malfunction. This lethal disease is caused by mutations in the OCRL1 gene, which encodes for the phosphatidylinositol 5-phosphatase Ocrl1. While in the past decade we witnessed substantial progress in the identification and characterization of LS patient cellular phenotypes, many of these studies have been performed in knocked-down cell lines or patient's cells from accessible cell types such as skin fibroblasts, and not from the organs affected. This is partially due to the limited accessibility of patient cells from eyes, brain and kidneys. Here we report the preparation of induced pluripotent stem cells (iPSCs) from patient skin fibroblasts and their reprogramming into kidney cells. These reprogrammed kidney cells displayed primary cilia assembly defects similar to those described previously in cell lines. Additionally, the transcription factor and cap mesenchyme marker Six2 was substantially retained in the Golgi complex and the functional nuclear-localized fraction was reduced. These results were confirmed using different batches of differentiated cells from different iPSC colonies and by the use of the human proximal tubule kidney cell line HK2. Indeed, OCRL1 KO led to both ciliogenesis defects and Six2 retention in the Golgi complex. In agreement with Six2's role in the suppression of ductal kidney lineages, cells from this pedigree were over-represented among patient kidney-reprogrammed cells. We speculate that this diminished efficacy to produce cap mesenchyme cells would cause LS patients to have difficulties in replenishing senescent or damaged cells derived from this lineage, particularly proximal tubule cells, leading to pathological scenarios such as tubular atrophy.
Collapse
Affiliation(s)
- Wen-Chieh Hsieh
- Department of Biological Sciences, Purdue University, West Lafayette, IN United States of America
| | - Swetha Ramadesikan
- Department of Biological Sciences, Purdue University, West Lafayette, IN United States of America
| | - Donna Fekete
- Department of Biological Sciences, Purdue University, West Lafayette, IN United States of America
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN United States of America
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN United States of America
- Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN United States of America
| | - Ruben Claudio Aguilar
- Department of Biological Sciences, Purdue University, West Lafayette, IN United States of America
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN United States of America
- Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN United States of America
- Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN United States of America
| |
Collapse
|
178
|
Muthukrishnan SD, Ryzhov S, Karolak M, Oxburgh L. Nephron progenitor cell death elicits a limited compensatory response associated with interstitial expansion in the neonatal kidney. Dis Model Mech 2018; 11:dmm.030544. [PMID: 29196442 PMCID: PMC5818074 DOI: 10.1242/dmm.030544] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 11/23/2017] [Indexed: 12/11/2022] Open
Abstract
The final nephron number in an adult kidney is regulated by nephron progenitor cell availability and collecting duct branching in the fetal period. Fetal environmental perturbations that cause reductions in cell numbers in these two compartments result in low nephron endowment. Previous work has shown that maternal dietary factors influence nephron progenitor cell availability, with both caloric restriction and protein deprivation leading to reduced cell numbers through apoptosis. In this study, we evaluate the consequences of inducing nephron progenitor cell death on progenitor niche dynamics and on nephron endowment. Depletion of approximately 40% of nephron progenitor cells by expression of diphtheria toxin A at embryonic day 15 in the mouse results in 10-20% nephron reduction in the neonatal period. Analysis of cell numbers within the progenitor cell pool following induction of apoptosis reveals a compensatory response in which surviving progenitor cells increase their proliferation and replenish the niche. The proliferative response is temporally associated with infiltration of macrophages into the nephrogenic zone. Colony stimulating factor 1 (CSF1) has a mitogenic effect on nephron progenitor cells, providing a potential explanation for the compensatory proliferation. However, CSF1 also promotes interstitial cell proliferation, and the compensatory response is associated with interstitial expansion in recovering kidneys which can be pharmacologically inhibited by treatment with clodronate liposomes. Our findings suggest that the fetal kidney employs a macrophage-dependent compensatory regenerative mechanism to respond to acute injury caused by death of nephron progenitor cells, but that this regenerative response is associated with neonatal interstitial expansion. Editor's choice: Formation of the kidney relies on maintaining progenitor cells throughout development. The authors find that apoptotic loss of nephron progenitor cells provokes compensatory proliferation mediated by trophic factors released by phagocytes.
Collapse
Affiliation(s)
- Sree Deepthi Muthukrishnan
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, USA.,Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA
| | - Sergey Ryzhov
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, USA
| | - Michele Karolak
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, USA
| | - Leif Oxburgh
- Center for Molecular Medicine, Maine Medical Center Research Institute, Scarborough, ME 04074, USA
| |
Collapse
|
179
|
Sanna-Cherchi S, Westland R, Ghiggeri GM, Gharavi AG. Genetic basis of human congenital anomalies of the kidney and urinary tract. J Clin Invest 2018; 128:4-15. [PMID: 29293093 DOI: 10.1172/jci95300] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The clinical spectrum of congenital anomalies of the kidney and urinary tract (CAKUT) encompasses a common birth defect in humans that has significant impact on long-term patient survival. Overall, data indicate that approximately 20% of patients may have a genetic disorder that is usually not detected based on standard clinical evaluation, implicating many different mutational mechanisms and pathogenic pathways. In particular, 10% to 15% of CAKUT patients harbor an unsuspected genomic disorder that increases risk of neurocognitive impairment and whose early recognition can impact clinical care. The emergence of high-throughput genomic technologies is expected to provide insight into the common and rare genetic determinants of diseases and offer opportunities for early diagnosis with genetic testing.
Collapse
Affiliation(s)
- Simone Sanna-Cherchi
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| | - Rik Westland
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA.,Department of Pediatric Nephrology, VU University Medical Center, Amsterdam, Netherlands
| | - Gian Marco Ghiggeri
- Division of Nephrology, Dialysis and Transplantation, Istituto Giannina Gaslini, Genoa, Italy
| | - Ali G Gharavi
- Division of Nephrology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, New York, USA
| |
Collapse
|
180
|
Puri P, Schaefer CM, Bushnell D, Taglienti ME, Kreidberg JA, Yoder BK, Bates CM. Ectopic Phosphorylated Creb Marks Dedifferentiated Proximal Tubules in Cystic Kidney Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2018; 188:84-94. [PMID: 29107072 PMCID: PMC5745541 DOI: 10.1016/j.ajpath.2017.09.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 08/18/2017] [Accepted: 09/11/2017] [Indexed: 01/25/2023]
Abstract
Ectopic cAMP signaling is pathologic in polycystic kidney disease; however, its spatiotemporal actions are unclear. We characterized the expression of phosphorylated Creb (p-Creb), a target and mediator of cAMP signaling, in developing and cystic kidney models. We also examined tubule-specific effects of cAMP analogs in cystogenesis in embryonic kidney explants. In wild-type mice, p-Creb marked nephron progenitors (NP), early epithelial NP derivatives, ureteric bud, and cortical stroma; p-Creb was present in differentiated thick ascending limb of Henle, collecting duct, and stroma; however, it disappeared in mature NP-derived proximal tubules. In Six2cre;Frs2αFl/Fl mice, a renal cystic model, ectopic p-Creb stained proximal tubule-derived cystic segments that lost the differentiation marker lotus tetragonolobus lectin. Furthermore, lotus tetragonolobus lectin-negative/p-Creb-positive cyst segments (re)-expressed Ncam1, Pax2, and Sox9 markers of immature nephron structures and dedifferentiated proximal tubules after acute kidney injury. These dedifferentiation markers were co-expressed with p-Creb in renal cysts in Itf88 knockout mice subjected to ischemia and Six2cre;Pkd1Fl/Fl mice, other renal cystogenesis models. 8-Br-cAMP addition to wild-type embryonic kidney explants induced proximal tubular cystogenesis and p-Creb expression; these effects were blocked by co-addition of protein kinase A inhibitor. Thus p-Creb/cAMP signaling is appropriate in NP and early nephron derivatives, but disappears in mature proximal tubules. Moreover, ectopic p-Creb expression/cAMP signaling marks dedifferentiated proximal tubular cystic segments. Furthermore, proximal tubules are predisposed to become cystic after cAMP stimulation.
Collapse
Affiliation(s)
- Pawan Puri
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Caitlin M Schaefer
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Daniel Bushnell
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Mary E Taglienti
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts; Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Jordan A Kreidberg
- Department of Urology, Boston Children's Hospital, Boston, Massachusetts; Department of Surgery, Harvard Medical School, Boston, Massachusetts
| | - Bradley K Yoder
- Department of Pediatrics, Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Carlton M Bates
- Division of Nephrology, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Division of Nephrology, Children's Hospital of Pittsburgh of University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
181
|
Roy Chowdhury N, Hopp I, Zilm P, Murray P, Vasilev K. Silver nanoparticle modified surfaces induce differentiation of mouse kidney-derived stem cells. RSC Adv 2018; 8:20334-20340. [PMID: 35541676 PMCID: PMC9080803 DOI: 10.1039/c8ra02145g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
In this paper, we interrogate the influence of silver nanoparticle (AgNPs)-based model surfaces on mouse kidney-derived stem cells (mKSCs) differentiation. The widespread use of silver in biomedical and consumer products requires understanding of this element's effect on kidney cells. Moreover, the potential for using stem cells in drug discovery require methods to direct their differentiation to specialized cells. Hence, we generated coated model substrates containing different concentrations of surface immobilized AgNPs, and used them to evaluate properties and functions of mKSCs. Initially, mKSCs exhibited reduced viability on higher silver containing surfaces. However, longer culture periods assisted mKSCs to recover. Greater degree of cell spreading and arborization led by AgNPs, suggest podocyte differentiation. Proximal tubule cell marker's expression revealed differentiation to the specific lineage. Although the exact mechanism underpinning these findings require significant future efforts, this study demonstrate silver's capacity to stimulate mKSC differentiation, which may provide opportunities for drug screenings. 2-Methyl-2-oxazoline plasma polymerized silver nanoparticles containing coatings are not toxic towards mouse kidney derived stem cells (mKSCs) and regulate mKSCs differentiation.![]()
Collapse
Affiliation(s)
| | - Isabel Hopp
- Institute of Translational Medicine
- University of Liverpool
- Liverpool
- UK
| | - Peter Zilm
- Microbiology Laboratory
- Adelaide Dental School
- The University of Adelaide
- Adelaide
- Australia
| | - Patricia Murray
- Institute of Translational Medicine
- University of Liverpool
- Liverpool
- UK
| | - Krasimir Vasilev
- School of Engineering
- University of South Australia
- Mawson Lakes
- Australia
| |
Collapse
|
182
|
Kobayashi H, Liu J, Urrutia AA, Burmakin M, Ishii K, Rajan M, Davidoff O, Saifudeen Z, Haase VH. Hypoxia-inducible factor prolyl-4-hydroxylation in FOXD1 lineage cells is essential for normal kidney development. Kidney Int 2017; 92:1370-1383. [PMID: 28847650 PMCID: PMC5696043 DOI: 10.1016/j.kint.2017.06.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 05/26/2017] [Accepted: 06/08/2017] [Indexed: 12/30/2022]
Abstract
Hypoxia in the embryo is a frequent cause of intra-uterine growth retardation, low birth weight, and multiple organ defects. In the kidney, this can lead to low nephron endowment, predisposing to chronic kidney disease and arterial hypertension. A key component in cellular adaptation to hypoxia is the hypoxia-inducible factor pathway, which is regulated by prolyl-4-hydroxylase domain (PHD) dioxygenases PHD1, PHD2, and PHD3. In the adult kidney, PHD oxygen sensors are differentially expressed in a cell type-dependent manner and control the production of erythropoietin in interstitial cells. However, the role of interstitial cell PHDs in renal development has not been examined. Here we used a genetic approach in mice to interrogate PHD function in FOXD1-expressing stroma during nephrogenesis. We demonstrate that PHD2 and PHD3 are essential for normal kidney development as the combined inactivation of stromal PHD2 and PHD3 resulted in renal failure that was associated with reduced kidney size, decreased numbers of glomeruli, and abnormal postnatal nephron formation. In contrast, nephrogenesis was normal in animals with individual PHD inactivation. We furthermore demonstrate that the defect in nephron formation in PHD2/PHD3 double mutants required intact hypoxia-inducible factor-2 signaling and was dependent on the extent of stromal hypoxia-inducible factor activation. Thus, hypoxia-inducible factor prolyl-4-hydroxylation in renal interstitial cells is critical for normal nephron formation.
Collapse
Affiliation(s)
- Hanako Kobayashi
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Jiao Liu
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University Health Sciences Center, New Orleans, Louisiana, USA; The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Andres A Urrutia
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Mikhail Burmakin
- Division of Vascular Biology, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Stockholm, Sweden
| | - Ken Ishii
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Malini Rajan
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | - Olena Davidoff
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Zubaida Saifudeen
- Section of Pediatric Nephrology, Department of Pediatrics, Tulane University Health Sciences Center, New Orleans, Louisiana, USA; The Hypertension and Renal Centers of Excellence, Tulane University Health Sciences Center, New Orleans, Louisiana, USA
| | - Volker H Haase
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; Medical and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee, USA; Department of Cancer Biology and Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee, USA.
| |
Collapse
|
183
|
De Tomasi L, David P, Humbert C, Silbermann F, Arrondel C, Tores F, Fouquet S, Desgrange A, Niel O, Bole-Feysot C, Nitschké P, Roume J, Cordier MP, Pietrement C, Isidor B, Khau Van Kien P, Gonzales M, Saint-Frison MH, Martinovic J, Novo R, Piard J, Cabrol C, Verma IC, Puri R, Journel H, Aziza J, Gavard L, Said-Menthon MH, Heidet L, Saunier S, Jeanpierre C. Mutations in GREB1L Cause Bilateral Kidney Agenesis in Humans and Mice. Am J Hum Genet 2017; 101:803-814. [PMID: 29100091 DOI: 10.1016/j.ajhg.2017.09.026] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/28/2017] [Indexed: 12/25/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) constitute a major cause of chronic kidney disease in children and 20% of prenatally detected anomalies. CAKUT encompass a spectrum of developmental kidney defects, including renal agenesis, hypoplasia, and cystic and non-cystic dysplasia. More than 50 genes have been reported as mutated in CAKUT-affected case subjects. However, the pathophysiological mechanisms leading to bilateral kidney agenesis (BKA) remain largely elusive. Whole-exome or targeted exome sequencing of 183 unrelated familial and/or severe CAKUT-affected case subjects, including 54 fetuses with BKA, led to the identification of 16 heterozygous variants in GREB1L (growth regulation by estrogen in breast cancer 1-like), a gene reported as a target of retinoic acid signaling. Four loss-of-function and 12 damaging missense variants, 14 being absent from GnomAD, were identified. Twelve of them were present in familial or simplex BKA-affected case subjects. Female BKA-affected fetuses also displayed uterus agenesis. We demonstrated a significant association between GREB1L variants and BKA. By in situ hybridization, we showed expression of Greb1l in the nephrogenic zone in developing mouse kidney. We generated a Greb1l knock-out mouse model by CRISPR-Cas9. Analysis at E13.5 revealed lack of kidneys and genital tract anomalies in male and female Greb1l-/- embryos and a slight decrease in ureteric bud branching in Greb1l+/- embryos. We showed that Greb1l invalidation in mIMCD3 cells affected tubulomorphogenesis in 3D-collagen culture, a phenotype rescued by expression of the wild-type human protein. This demonstrates that GREB1L plays a major role in early metanephros and genital development in mice and humans.
Collapse
|
184
|
Sequeira-Lopez MLS, Torban E. New insights into precursors of renal endothelium. Kidney Int 2017; 90:244-246. [PMID: 27418087 DOI: 10.1016/j.kint.2016.03.043] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/23/2016] [Indexed: 02/01/2023]
Abstract
The kidney vasculature is extremely complex, yet, despite recent progress, our understanding of how the renal vascular system develops is limited. By using advanced tissue engineering techniques and in vivo and in vitro depletion of specific populations of endothelial cell precursors, Halt et al. have identified a CD146-expressing precursor as an important player in the development of the renal vasculature.
Collapse
Affiliation(s)
| | - Elena Torban
- McGill University and McGill University Health Center Research Institute, Department of Medicine/Nephrology, Montreal, Quebec, Canada.
| |
Collapse
|
185
|
Fukuzawa R, Anaka MR, Morison IM, Reeve AE. The developmental programme for genesis of the entire kidney is recapitulated in Wilms tumour. PLoS One 2017; 12:e0186333. [PMID: 29040332 PMCID: PMC5645110 DOI: 10.1371/journal.pone.0186333] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 10/01/2017] [Indexed: 11/19/2022] Open
Abstract
Wilms tumour (WT) is an embryonal tumour that recapitulates kidney development. The normal kidney is formed from two distinct embryological origins: the metanephric mesenchyme (MM) and the ureteric bud (UB). It is generally accepted that WT arises from precursor cells in the MM; however whether UB-equivalent structures participate in tumorigenesis is uncertain. To address the question of the involvement of UB, we assessed 55 Wilms tumours for the molecular features of MM and UB using gene expression profiling, immunohistochemsitry and immunofluorescence. Expression profiling primarily based on the Genitourinary Molecular Anatomy Project data identified molecular signatures of the UB and collecting duct as well as those of the proximal and distal tubules in the triphasic histology group. We performed immunolabeling for fetal kidneys and WTs. We focused on a central epithelial blastema pattern which is the characteristic of triphasic histology characterized by UB-like epithelial structures surrounded by MM and MM-derived epithelial structures, evoking the induction/aggregation phase of the developing kidney. The UB-like epithelial structures and surrounding MM and epithelial structures resembling early glomerular epithelium, proximal and distal tubules showed similar expression patterns to those of the developing kidney. These observations indicate WTs can arise from a precursor cell capable of generating the entire kidney, such as the cells of the intermediate mesoderm from which both the MM and UB are derived. Moreover, this provides an explanation for the variable histological features of mesenchymal to epithelial differentiation seen in WT.
Collapse
Affiliation(s)
- Ryuji Fukuzawa
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Department of Pathology, Tokyo Metropolitan Children's Medical Center, Fuchu, Japan
- Department of Pathology, University of Otago, Dunedin, New Zealand
- Department of Pathology, School of Medicine, International University of Health and Welfare, Narita, Japan
- * E-mail:
| | - Matthew R. Anaka
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin, New Zealand
- Department of Medicine, University of Toronto, Toronto, Canada
| | - Ian M. Morison
- Department of Pathology, University of Otago, Dunedin, New Zealand
| | - Anthony E. Reeve
- Cancer Genetics Laboratory, Department of Biochemistry, University of Otago, Dunedin, New Zealand
| |
Collapse
|
186
|
Pan X, Karner CM, Carroll TJ. Myc cooperates with β-catenin to drive gene expression in nephron progenitor cells. Development 2017; 144:4173-4182. [PMID: 28993399 DOI: 10.1242/dev.153700] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 10/03/2017] [Indexed: 12/19/2022]
Abstract
For organs to achieve their proper size, the processes of stem cell renewal and differentiation must be tightly regulated. We previously showed that in the developing kidney, Wnt9b regulates distinct β-catenin-dependent transcriptional programs in the renewing and differentiating populations of the nephron progenitor cells. How β-catenin stimulated these two distinct programs was unclear. Here, we show that β-catenin cooperates with the transcription factor Myc to activate the progenitor renewal program. Although in multiple contexts Myc is a target of β-catenin, our characterization of a cell type-specific enhancer for the Wnt9b/β-catenin target gene Fam19a5 shows that Myc and β-catenin cooperate to activate gene expression controlled by this element. This appears to be a more general phenomenon as we find that Myc is required for the expression of every Wnt9b/β-catenin progenitor renewal target assessed as well as for proper nephron endowment in vivo This study suggests that, within the developing kidney, tissue-specific β-catenin activity is regulated by cooperation with cell type-specific transcription factors. This finding not only provides insight into the regulation of β-catenin target genes in the developing kidney, but will also advance our understanding of progenitor cell renewal in other cell types/organ systems in which Myc and β-catenin are co-expressed.
Collapse
Affiliation(s)
- Xinchao Pan
- Department of Internal Medicine (Nephrology), UT Southwestern Medical Center, Dallas, TX 75390-9148, USA.,Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA
| | - Courtney M Karner
- Department of Internal Medicine (Nephrology), UT Southwestern Medical Center, Dallas, TX 75390-9148, USA.,Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA.,Department of Orthopaedic Surgery and Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Thomas J Carroll
- Department of Internal Medicine (Nephrology), UT Southwestern Medical Center, Dallas, TX 75390-9148, USA .,Molecular Biology, UT Southwestern Medical Center, Dallas, TX 75390-9148, USA
| |
Collapse
|
187
|
Kaverina NV, Eng DG, Largent AD, Daehn I, Chang A, Gross KW, Pippin JW, Hohenstein P, Shankland SJ. WT1 Is Necessary for the Proliferation and Migration of Cells of Renin Lineage Following Kidney Podocyte Depletion. Stem Cell Reports 2017; 9:1152-1166. [PMID: 28966119 PMCID: PMC5639431 DOI: 10.1016/j.stemcr.2017.08.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/17/2022] Open
Abstract
Wilms' tumor suppressor 1 (WT1) plays an important role in cell proliferation and mesenchymal-epithelial balance in normal development and disease. Here, we show that following podocyte depletion in three experimental models, and in patients with focal segmental glomerulosclerosis (FSGS) and membranous nephropathy, WT1 increased significantly in cells of renin lineage (CoRL). In an animal model of FSGS in RenWt1fl/fl reporter mice with inducible deletion of WT1 in CoRL, CoRL proliferation and migration to the glomerulus was reduced, and glomerular disease was worse compared with wild-type mice. To become podocytes, CoRL undergo mesenchymal-to-epithelial transformation (MET), typified by reduced staining for mesenchymal markers (MYH11, SM22, αSMA) and de novo expression of epithelial markers (E-cadherin and cytokeratin18). Evidence for changes in MET markers was barely detected in RenWt1fl/fl mice. Our results show that following podocyte depletion, WT1 plays essential roles in CoRL proliferation and migration toward an adult podocyte fate.
Collapse
Affiliation(s)
- Natalya V Kaverina
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Diana G Eng
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Andrea D Largent
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Ilse Daehn
- Department of Medicine, Division of Nephrology, The Icahn School of Medicine at Mount Sinai, 1468 Madison Avenue, New York, NY 10029, USA
| | - Anthony Chang
- Department of Pathology, University of Chicago, 5841 S Maryland Ave, Chicago, IL 60637, USA
| | - Kenneth W Gross
- Department of Molecular and Cellular Biology, Roswell Park Cancer Institute, Elm & Carlton Streets, Buffalo, NY 14263, USA
| | - Jeffrey W Pippin
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush, Midlothian EH25 9RG, UK
| | - Stuart J Shankland
- Division of Nephrology, University of Washington School of Medicine, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
188
|
Schmidt-Ott KM. How to grow a kidney: patient-specific kidney organoids come of age. Nephrol Dial Transplant 2017; 32:17-23. [PMID: 27411722 DOI: 10.1093/ndt/gfw256] [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: 04/21/2016] [Accepted: 06/02/2016] [Indexed: 12/23/2022] Open
Abstract
The notion of regrowing a patient's kidney in a dish has fascinated researchers for decades and has spurred visions of revolutionary clinical applications. Recently, this option has come closer to reality. Key technologies have been developed to generate patient-specific pluripotent stem cells and to edit their genome. Several laboratories have devised protocols to differentiate patient-specific pluripotent stem cells into kidney cells or into in vitro organoids that resemble the kidney with respect to cell types, tissue architecture and disease pathology. This was possible because of rapidly expanding knowledge regarding the cellular and molecular basis of embryonic kidney development. Generating kidney cells or organoids from patient-specific stem cells may prove to be clinically useful in several ways. First, patient-specific kidney cells or organoids could be used to predict an individual's response to stressors, toxins or medications and thereby develop personalized treatment decisions. Second, patient-specific stem cells harbour the individual's genetic defects. This may potentially enable genetic rescue attempts to establish the significance of a genetic defect in a stem cell-derived organoid or it may allow testing of patient-specific targeted therapies for kidney disease in vitro. From a tissue engineering perspective, patient-specific kidney organoids might provide a key advance towards engineering immunocompatible transplantable kidneys. This review article summarizes recent developments in the field and discusses its current limitations and future perspectives.
Collapse
Affiliation(s)
- Kai M Schmidt-Ott
- Department of Nephrology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Molecular and Translational Kidney Research, Max Delbrueck Center for Molecular Medicine, Berlin, Germany
| |
Collapse
|
189
|
El-Dahr S, Hilliard S, Saifudeen Z. Regulation of kidney development by the Mdm2/Mdm4-p53 axis. J Mol Cell Biol 2017; 9:26-33. [PMID: 28096292 PMCID: PMC5907835 DOI: 10.1093/jmcb/mjx005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/15/2017] [Indexed: 01/13/2023] Open
Abstract
While p53 activity is required for tumour suppression, unconstrained p53 activity on the other hand is detrimental to the organism, resulting in inappropriate cellular death or proliferation defects. Unimpeded p53 activity is lethal in the developing embryo, underlining the need for maintaining a tight control on p53 activity during this period. The critical role of the negative regulators of p53, Mdm2 and Mdm4, in vertebrate development came to light by fatal disruption of embryogenesis that was observed with Mdm2 and Mdm4 gene deletions in mice. Embryonic lethality was rescued only by superimposing p53 removal. Here we summarize the contribution of the Mdm2/Mdm4–p53 axis that occurs at multiple steps of kidney development. Conditional, cell type-specific deletions reveal distinct functions of these proteins in renal morphogenesis. The severe impact on the renal phenotype from targeted gene deletions underscores the critical role played by the Mdm2/Mdm4–p53 nexus on nephrogenesis, and emphasizes the need to monitor patients with aberrations in this pathway for kidney function defects and associated cardiovascular dysfunction.
Collapse
Affiliation(s)
- Samir El-Dahr
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Sylvia Hilliard
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Zubaida Saifudeen
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
190
|
Naiman N, Fujioka K, Fujino M, Valerius MT, Potter SS, McMahon AP, Kobayashi A. Repression of Interstitial Identity in Nephron Progenitor Cells by Pax2 Establishes the Nephron-Interstitium Boundary during Kidney Development. Dev Cell 2017; 41:349-365.e3. [PMID: 28535371 DOI: 10.1016/j.devcel.2017.04.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 03/10/2017] [Accepted: 04/24/2017] [Indexed: 12/31/2022]
Abstract
The kidney contains the functional units, the nephrons, surrounded by the renal interstitium. Previously we discovered that, once Six2-expressing nephron progenitor cells and Foxd1-expressing renal interstitial progenitor cells form at the onset of kidney development, descendant cells from these populations contribute exclusively to the main body of nephrons and renal interstitial tissues, respectively, indicating a lineage boundary between the nephron and renal interstitial compartments. Currently it is unclear how lineages are regulated during kidney organogenesis. We demonstrate that nephron progenitor cells lacking Pax2 fail to differentiate into nephron cells but can switch fates into renal interstitium-like cell types. These data suggest that Pax2 function maintains nephron progenitor cells by repressing a renal interstitial cell program. Thus, the lineage boundary between the nephron and renal interstitial compartments is maintained by the Pax2 activity in nephron progenitor cells during kidney organogenesis.
Collapse
Affiliation(s)
- Natalie Naiman
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Kaoru Fujioka
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Mari Fujino
- Department of Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, 750 Republican Street, Seattle, WA 98109, USA
| | - M Todd Valerius
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Medical Center, 3333 Burnet Avenue, Cincinnati, OH 45229, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, 1425 San Pablo Street, Los Angeles, CA 90033, USA
| | - Akio Kobayashi
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA; Department of Medicine, Institute for Stem Cell and Regenerative Medicine, University of Washington, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
191
|
Liu J, Edgington-Giordano F, Dugas C, Abrams A, Katakam P, Satou R, Saifudeen Z. Regulation of Nephron Progenitor Cell Self-Renewal by Intermediary Metabolism. J Am Soc Nephrol 2017; 28:3323-3335. [PMID: 28754792 DOI: 10.1681/asn.2016111246] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 06/09/2017] [Indexed: 12/21/2022] Open
Abstract
Nephron progenitor cells (NPCs) show an age-dependent capacity to balance self-renewal with differentiation. Older NPCs (postnatal day 0) exit the progenitor niche at a higher rate than younger (embryonic day 13.5) NPCs do. This behavior is reflected in the transcript profiles of young and old NPCs. Bioenergetic pathways have emerged as important regulators of stem cell fate. Here, we investigated the mechanisms underlying this regulation in murine NPCs. Upon isolation and culture in NPC renewal medium, younger NPCs displayed a higher glycolysis rate than older NPCs. Inhibition of glycolysis enhanced nephrogenesis in cultured embryonic kidneys, without increasing ureteric tree branching, and promoted mesenchymal-to-epithelial transition in cultured isolated metanephric mesenchyme. Cotreatment with a canonical Wnt signaling inhibitor attenuated but did not entirely block the increase in nephrogenesis observed after glycolysis inhibition. Furthermore, inhibition of the phosphatidylinositol 3-kinase/Akt self-renewal signaling pathway or stimulation of differentiation pathways in the NPC decreased glycolytic flux. Our findings suggest that glycolysis is a pivotal, cell-intrinsic determinant of NPC fate, with a high glycolytic flux supporting self-renewal and inhibition of glycolysis stimulating differentiation.
Collapse
Affiliation(s)
- Jiao Liu
- Department of Pediatrics, Section of Nephrology
| | | | | | - Anna Abrams
- Department of Pediatrics, Section of Nephrology
| | - Prasad Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, Louisiana
| | | | | |
Collapse
|
192
|
Abstract
p53 is best identified as a tumor suppressor for its transcriptional control of genes involved in cell cycle progression and apoptosis. Beyond its irrefutable involvement in restraining unchecked cell proliferation, research over the past several years has indicated a requirement for p53 function in sustaining normal development. Here I summarize the role of p53 in embryonic development, with a focus on knowledge gained from p53 loss and overexpression during kidney development. In contrast to its classical role in suppressing proliferative pathways, p53 positively regulates nephron progenitor cell (NPC) renewal. Emerging evidence suggests p53 may control cell fate decisions by preserving energy metabolism homeostasis of progenitors in the nephrogenic niche. Maintaining a critical level of p53 function appears to be a prerequisite for optimal nephron endowment. Defining the molecular networks targeted by p53 in the NPC may well provide new targets not only for regenerative medicine but also for cancer treatment.
Collapse
Affiliation(s)
- Zubaida Saifudeen
- Department of Pediatrics, Section of Pediatric Nephrology, Tulane University School of Medicine, 1430 Tulane Avenue, SL37, New Orleans, LA, 70112, USA.
| |
Collapse
|
193
|
Chen L, Higgins PJ, Zhang W. Development and Diseases of the Collecting Duct System. Results Probl Cell Differ 2017; 60:165-203. [PMID: 28409346 DOI: 10.1007/978-3-319-51436-9_7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The collecting duct of the mammalian kidney is important for the regulation of extracellular volume, osmolarity, and pH. There are two major structurally and functionally distinct cell types: principal cells and intercalated cells. The former regulates Na+ and water homeostasis, while the latter participates in acid-base homeostasis. In vivo lineage tracing using Cre recombinase or its derivatives such as CreGFP and CreERT2 is a powerful new technique to identify stem/progenitor cells in their native environment and to decipher the origins of the tissue that they give rise to. Recent studies using this technique in mice have revealed multiple renal progenitor cell populations that differentiate into various nephron segments and collecting duct. In particular, emerging evidence suggests that like principal cells, most of intercalated cells originate from the progenitor cells expressing water channel Aquaporin 2. Mutations or malfunctions of the channels, pumps, and transporters expressed in the collecting duct system cause various human diseases. For example, gain-of-function mutations in ENaC cause Liddle's syndrome, while loss-of-function mutations in ENaC lead to Pseudohypoaldosteronism type 1. Mutations in either AE1 or V-ATPase B1 result in distal renal tubular acidosis. Patients with disrupted AQP2 or AVPR2 develop nephrogenic diabetes insipidus. A better understanding of the function and development of the collecting duct system may facilitate the discovery of new therapeutic strategies for treating kidney disease.
Collapse
Affiliation(s)
- Lihe Chen
- Epithelial Systems Biology Laboratory, Systems Biology Center, NHLBI, Bethesda, MD, 20892-1603, USA
| | - Paul J Higgins
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Wenzheng Zhang
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| |
Collapse
|
194
|
Rutledge EA, Benazet JD, McMahon AP. Cellular heterogeneity in the ureteric progenitor niche and distinct profiles of branching morphogenesis in organ development. Development 2017; 144:3177-3188. [PMID: 28705898 DOI: 10.1242/dev.149112] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 07/10/2017] [Indexed: 12/26/2022]
Abstract
Branching morphogenesis creates arborized epithelial networks. In the mammalian kidney, an epithelial progenitor pool at ureteric branch tips (UBTs) creates the urine-transporting collecting system. Using region-specific mouse reporter strains, we performed an RNA-seq screen, identifying tip- and stalk-enriched gene sets in the developing collecting duct system. Detailed in situ hybridization studies of tip-enriched predictions identified UBT-enriched gene sets conserved between the mouse and human kidney. Comparative spatial analysis of their UBT niche expression highlighted distinct patterns of gene expression revealing novel molecular heterogeneity within the UBT progenitor population. To identify kidney-specific and shared programs of branching morphogenesis, comparative expression studies on the developing mouse lung were combined with in silico analysis of the developing mouse salivary gland. These studies highlight a shared gene set with multi-organ tip enrichment and a gene set specific to UBTs. This comprehensive analysis extends our current understanding of the ureteric branch tip niche.
Collapse
Affiliation(s)
- Elisabeth A Rutledge
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| | - Jean-Denis Benazet
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA.,Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, CA 94143, USA
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
195
|
Werth M, Schmidt-Ott KM, Leete T, Qiu A, Hinze C, Viltard M, Paragas N, Shawber CJ, Yu W, Lee P, Chen X, Sarkar A, Mu W, Rittenberg A, Lin CS, Kitajewski J, Al-Awqati Q, Barasch J. Transcription factor TFCP2L1 patterns cells in the mouse kidney collecting ducts. eLife 2017; 6. [PMID: 28577314 PMCID: PMC5484618 DOI: 10.7554/elife.24265] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 06/03/2017] [Indexed: 12/19/2022] Open
Abstract
Although most nephron segments contain one type of epithelial cell, the collecting ducts consists of at least two: intercalated (IC) and principal (PC) cells, which regulate acid-base and salt-water homeostasis, respectively. In adult kidneys, these cells are organized in rosettes suggesting functional interactions. Genetic studies in mouse revealed that transcription factor Tfcp2l1 coordinates IC and PC development. Tfcp2l1 induces the expression of IC specific genes, including specific H+-ATPase subunits and Jag1. Jag1 in turn, initiates Notch signaling in PCs but inhibits Notch signaling in ICs. Tfcp2l1 inactivation deletes ICs, whereas Jag1 inactivation results in the forfeiture of discrete IC and PC identities. Thus, Tfcp2l1 is a critical regulator of IC-PC patterning, acting cell-autonomously in ICs, and non-cell-autonomously in PCs. As a result, Tfcp2l1 regulates the diversification of cell types which is the central characteristic of 'salt and pepper' epithelia and distinguishes the collecting duct from all other nephron segments. DOI:http://dx.doi.org/10.7554/eLife.24265.001
Collapse
Affiliation(s)
- Max Werth
- Columbia University, New York, United States
| | - Kai M Schmidt-Ott
- Columbia University, New York, United States.,Max Delbruck Center for Molecular Medicine, Berlin, Germany.,Department of Nephrology and Intensive Care Medicine, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | | | - Andong Qiu
- Columbia University, New York, United States.,Tongji University, Shanghai, China
| | | | - Melanie Viltard
- Columbia University, New York, United States.,Institute for European Expertise in Physiology, Paris, France
| | - Neal Paragas
- Columbia University, New York, United States.,University of Washington, Seattle, United States
| | | | - Wenqiang Yu
- Columbia University, New York, United States.,Fudan University, Shanghai, China
| | - Peter Lee
- Columbia University, New York, United States
| | - Xia Chen
- Columbia University, New York, United States
| | - Abby Sarkar
- Columbia University, New York, United States
| | - Weiyi Mu
- Columbia University, New York, United States
| | | | | | - Jan Kitajewski
- Columbia University, New York, United States.,University of Illinois at Chicago, Chicago, United States
| | | | | |
Collapse
|
196
|
Balasubramaniam SL, Gopalakrishnapillai A, Petrelli NJ, Barwe SP. Knockdown of sodium-calcium exchanger 1 induces epithelial-to-mesenchymal transition in kidney epithelial cells. J Biol Chem 2017; 292:11388-11399. [PMID: 28550085 DOI: 10.1074/jbc.m116.752352] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 05/17/2017] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal-to-epithelial transition (MET) and epithelial-to-mesenchymal transition (EMT) are important processes in kidney development. Failure to undergo MET during development leads to the initiation of Wilms tumor, whereas EMT contributes to the development of renal cell carcinomas (RCC). The role of calcium regulators in governing these processes is becoming evident. We demonstrated earlier that Na+/Ca2+ exchanger 1 (NCX1), a major calcium exporter in renal epithelial cells, regulates epithelial cell motility. Here, we show for the first time that NCX1 mRNA and protein expression was down-regulated in Wilms tumor and RCC. Knockdown of NCX1 in Madin-Darby canine kidney cells induced fibroblastic morphology, increased intercellular junctional distance, and induced paracellular permeability, loss of apico-basal polarity in 3D cultures, and anchorage-independent growth, accompanied by expression of mesenchymal markers. We also provide evidence that NCX1 interacts with and anchors E-cadherin to the cell surface independent of NCX1 ion transport activity. Consistent with destabilization of E-cadherin, NCX1 knockdown cells showed an increase in β-catenin nuclear localization, enhanced transcriptional activity, and up-regulation of downstream targets of the β-catenin signaling pathway. Taken together, knockdown of NCX1 in Madin-Darby canine kidney cells alters epithelial morphology and characteristics by destabilization of E-cadherin and induction of β-catenin signaling.
Collapse
Affiliation(s)
- Sona Lakshme Balasubramaniam
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803.,the Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, and
| | - Anilkumar Gopalakrishnapillai
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803
| | - Nicholas J Petrelli
- the Helen F. Graham Cancer Center, Christiana Care Health System, Newark, Delaware 19718
| | - Sonali P Barwe
- From the Nemours Center for Childhood Cancer Research, Alfred I. duPont Hospital for Children, Wilmington, Delaware 19803, .,the Department of Biological Sciences, University of Delaware, Newark, Delaware 19716, and
| |
Collapse
|
197
|
Abstract
Technical advances in the development of organoid systems enable cell lines, primary adult cells, or stem or progenitor cells to develop into diverse, multicellular entities, which can self-renew, self-organize, and differentiate. These 3D organoid cultures have proven to be of value in increasing our understanding of the biology of disease and offer the potential of regenerative and genetic therapies. The successful application of 3D organoids derived from adult tissue into urological cancer research can further our understanding of these diseases and could also provide preclinical cancer models to realize the precision medicine paradigm by therapeutic screening of individual patient samples ex vivo. Kidney organoids derived from induced pluripotent stem cells provide personalized biomarkers, which can be correlated with genetic and clinical information. Organoid models can also improve our comprehension of aspects of particular diseases; for example, in prostate cancer, 3D organoids can aid in the identification of tumour-initiating cells from an epithelial cell lineage. Furthermore, kidney organoid differentiation from human pluripotent stem cells enables gene editing to model disease in kidney tubular epithelial cells. State-of-the-art human organoid cultures have potential as tools in basic and clinical research in renal, bladder, and prostatic diseases.
Collapse
Affiliation(s)
- Shangqian Wang
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Urology Department, the First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Dong Gao
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Key Laboratory of Systems Biology,CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, 320 Yueyang Road, Shanghai 200031, China
| | - Yu Chen
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, USA.,Genitourinary Oncology Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, New York 10065, USA.,Department of Cell and Developmental Biology, Weill Cornell Medical College, 1300 York Avenue, New York, New York 10065, USA
| |
Collapse
|
198
|
TβRII Regulates the Proliferation of Metanephric Mesenchyme Cells through Six2 In Vitro. Int J Mol Sci 2017; 18:ijms18040853. [PMID: 28420207 PMCID: PMC5412437 DOI: 10.3390/ijms18040853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 03/22/2017] [Accepted: 04/11/2017] [Indexed: 12/12/2022] Open
Abstract
The transforming growth factor-β (TGFβ) family signaling pathways play an important role in regulatory cellular networks and exert specific effects on developmental programs during embryo development. However, the function of TGFβ signaling pathways on the early kidney development remains unclear. In this work, we aim to detect the underlying role of TGFβ type II receptor (TβRII) in vitro, which has a similar expression pattern as the crucial regulator Six2 during early kidney development. Firstly, the 5-ethynyl-2′-deoxyuridine (EdU) assay showed knock down of TβRII significantly decreased the proliferation ratio of metanephric mesenchyme (MM) cells. Additionally, real-time Polymerase Chain Reaction (PCR) and Western blot together with immunofluorescence determined that the mRNA and protein levels of Six2 declined after TβRII knock down. Also, Six2 was observed to be able to partially rescue the proliferation phenotype caused by the depletion of TβRII. Moreover, bioinformatics analysis and luciferase assay indicated Smad3 could transcriptionally target Six2. Further, the EdU assay showed that Smad3 could also rescue the inhibition of proliferation caused by the knock down of TβRII. Taken together, these findings delineate the important function of the TGFβ signaling pathway in the early development of kidney and TβRII was shown to be able to promote the expression of Six2 through Smad3 mediating transcriptional regulation and in turn activate the proliferation of MM cells.
Collapse
|
199
|
Abstract
The success of anticancer therapy is usually limited by the development of drug resistance. Such acquired resistance is driven, in part, by intratumoural heterogeneity - that is, the phenotypic diversity of cancer cells co-inhabiting a single tumour mass. The introduction of the cancer stem cell (CSC) concept, which posits the presence of minor subpopulations of CSCs that are uniquely capable of seeding new tumours, has provided a framework for understanding one dimension of intratumoural heterogeneity. This concept, taken together with the identification of the epithelial-to-mesenchymal transition (EMT) programme as a critical regulator of the CSC phenotype, offers an opportunity to investigate the nature of intratumoural heterogeneity and a possible mechanistic basis for anticancer drug resistance. In fact, accumulating evidence indicates that conventional therapies often fail to eradicate carcinoma cells that have entered the CSC state via activation of the EMT programme, thereby permitting CSC-mediated clinical relapse. In this Review, we summarize our current understanding of the link between the EMT programme and the CSC state, and also discuss how this knowledge can contribute to improvements in clinical practice.
Collapse
|
200
|
Trionfini P, Benigni A. MicroRNAs as Master Regulators of Glomerular Function in Health and Disease. J Am Soc Nephrol 2017; 28:1686-1696. [PMID: 28232619 DOI: 10.1681/asn.2016101117] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are important regulators of gene expression, and the dysregulation of miRNAs is a common feature of several diseases. More miRNAs are identified almost daily, revealing the complexity of these transcripts in eukaryotic cellular networks. The study of renal miRNAs, using genetically modified mice or by perturbing endogenous miRNA levels, has revealed the important biologic roles miRNAs have in the major cell lineages that compose the glomerulus. Here, we provide an overview of miRNA biogenesis and function in regulating key genes and cellular pathways in glomerular cells during development and homeostasis. Moreover, we focus on the emerging mechanisms through which miRNAs contribute to different diseases affecting the glomerulus, such as FSGS, IgA nephropathy, lupus nephritis, and diabetic nephropathy. In-depth knowledge of miRNA-based gene regulation has made it possible to unravel pathomechanisms, enabling the design of new therapeutic strategies for glomerular diseases for which available therapies are not fully efficacious.
Collapse
Affiliation(s)
- Piera Trionfini
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| | - Ariela Benigni
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Centro Anna Maria Astori, Science and Technology Park Kilometro Rosso, Bergamo, Italy
| |
Collapse
|