1
|
Devlin LA, Dewhurst RM, Sudhindar PD, Sayer JA. Renal ciliopathies. Curr Top Dev Biol 2025; 163:229-305. [PMID: 40254346 DOI: 10.1016/bs.ctdb.2025.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Primary cilia are essential cellular organelles with pivotal roles in many signalling pathways. Here we provide an overview of the role of primary cilia within the kidney, starting with primary ciliary structure and key protein complexes. We then highlight the specialised functions of primary cilia, emphasising their role in a group of diseases known as renal ciliopathies. These conditions include forms of polycystic kidney disease, nephronophthisis, and other syndromic ciliopathies, such as Joubert syndrome and Bardet-Biedl syndrome. We explore models of renal ciliopathies, both in vitro and in vivo, shedding light on the molecular mechanisms underlying these diseases including Wnt and Hedgehog signalling pathways, inflammation, and cellular metabolism. Finally, we discuss therapeutic approaches, from current treatments to cutting-edge preclinical research and clinical trials.
Collapse
Affiliation(s)
- Laura A Devlin
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca M Dewhurst
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Praveen D Sudhindar
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - John A Sayer
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Renal Services, Newcastle upon Tyne NHS Foundation Trust, Newcastle upon Tyne, United Kingdom; National Institute for Health Research, Newcastle Biomedical Research Centre, Newcastle Upon Tyne, United Kingdom.
| |
Collapse
|
2
|
Wiegel J, Helmstädter M, Walz G, Bergen MD. Spontaneous Calcium Bursts Organize the Apical Actin Cytoskeleton of Multiciliated Cells. Int J Mol Sci 2025; 26:2507. [PMID: 40141151 PMCID: PMC11942550 DOI: 10.3390/ijms26062507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/05/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Motile cilia perform crucial functions during embryonic development and in adult tissues. They are anchored by an apical actin network that forms microridge-like structures on the surface of multiciliated cells. Using Xenopus as a model system to investigate the mechanisms underlying the formation of these specialized actin structures, we observed stochastic bursts of intracellular calcium concentration in developing multiciliated cells. Through optogenetic manipulation of calcium signaling, we found that individual calcium bursts triggered the fusion and extension of actin structures by activating non-muscle myosin. Repeated cycles of calcium activation promoted assembly and coherence of the maturing apical actin network. Inhibition of the endogenous inositol triphosphate-calcium pathway disrupted the formation of apical actin/microridge-like structures by reducing local centriolar RhoA signaling. This disruption was rescued by transient expression of constitutively active RhoA in multiciliated cells. Our findings identify repetitive calcium bursts as a driving force that promotes the self-organization of the highly specialized actin cytoskeleton of multiciliated cells.
Collapse
Affiliation(s)
- Johannes Wiegel
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
| | - Martin Helmstädter
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
- EMcore, Renal Division, Department of Medicine, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
- BIOSS and CIBSS Centre for Integrative Biological Signalling, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany
| | - Max D. Bergen
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106 Freiburg, Germany; (J.W.); (M.H.); (G.W.)
| |
Collapse
|
3
|
Wang H, Zaiser F, Eckert P, Ruf J, Kayser N, Veenstra AC, Müller M, Haas R, Walz G, Yakulov TA. Inversin (NPHP2) and Vangl2 are required for normal zebrafish cloaca formation. Biochem Biophys Res Commun 2023; 673:9-15. [PMID: 37352572 DOI: 10.1016/j.bbrc.2023.06.058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 05/31/2023] [Accepted: 06/17/2023] [Indexed: 06/25/2023]
Abstract
Nephronophthisis (NPH), an autosomal recessive ciliopathy, results from mutations in more than 20 different genes (NPHPs). These gene products form protein complexes that regulate trafficking within the cilium, a microtubular structure that plays a crucial role in developmental processes. Several NPHPs, including NPHP2/Inversin, have been linked to extraciliary functions. In addition to defining a specific segment of primary cilia (Inversin compartment), NPHP2 participates in planar cell polarity (PCP) signaling along with Dishevelled and Vangl family members. We used the mutant zebrafish line invssa36157, containing a stop codon at amino acid 314, to characterize tissue-specific functions of zebrafish Nphp2. The invssa36157 line exhibits mild ciliopathy phenotypes and increased glomerular and cloaca cyst formation. These mutants showed enhanced susceptibility to the simultaneous depletion of the nphp1/nphp2/nphp8 module, known to be involved in the cytoskeletal organization of epithelial cells. Notably, simultaneous depletion of zebrafish nphp1 and vangl2 led to a pronounced increase in cloaca malformations in the invssa36157 mutant embryos. Time-lapse imaging showed that the pronephric cells correctly migrated towards the ectodermal cells in these embryos, but failed to form the cloaca opening. Despite these abnormal developments, cellular fate does not seem to be affected in nphp1 and vangl2 MO-depleted invssa36157 mutants, as shown by in situ hybridizations for markers of pronephros and ectodermal cell development. However, significantly reduced apoptotic activity was observed in this double knockdown model, signifying the role of apoptosis in cloacal morphogenesis. Our findings underscore the critical interplay of nphp1, nphp2/Inversin, and vangl2 in orchestrating normal cloaca formation in zebrafish, shedding light on the complex molecular mechanisms underlying ciliopathy-associated phenotypes.
Collapse
Affiliation(s)
- Hui Wang
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Friedemann Zaiser
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Priska Eckert
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Johannes Ruf
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Nicolas Kayser
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Anna C Veenstra
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Merle Müller
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Rebecca Haas
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany
| | - Gerd Walz
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Albertstrasse 19, 79104, Freiburg, Germany
| | - Toma A Yakulov
- Renal Division, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Germany.
| |
Collapse
|
4
|
Grand K, Stoltz M, Rizzo L, Röck R, Kaminski MM, Salinas G, Getwan M, Naert T, Pichler R, Lienkamp SS. HNF1B Alters an Evolutionarily Conserved Nephrogenic Program of Target Genes. J Am Soc Nephrol 2023; 34:412-432. [PMID: 36522156 PMCID: PMC10103355 DOI: 10.1681/asn.2022010076] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 12/23/2022] Open
Abstract
SIGNIFICANCE STATEMENT Mutations in hepatocyte nuclear factor-1 β ( HNF1B ) are the most common monogenic causes of congenital renal malformations. HNF1B is necessary to directly reprogram fibroblasts to induced renal tubule epithelial cells (iRECs) and, as we demonstrate, can induce ectopic pronephric tissue in Xenopus ectodermal organoids. Using these two systems, we analyzed the effect of HNF1B mutations found in patients with cystic dysplastic kidney disease. We found cross-species conserved targets of HNF1B, identified transcripts that are differentially regulated by the patient-specific mutant protein, and functionally validated novel HNF1B targets in vivo . These results highlight evolutionarily conserved transcriptional mechanisms and provide insights into the genetic circuitry of nephrogenesis. BACKGROUND Hepatocyte nuclear factor-1 β (HNF1B) is an essential transcription factor during embryogenesis. Mutations in HNF1B are the most common monogenic causes of congenital cystic dysplastic renal malformations. The direct functional consequences of mutations in HNF1B on its transcriptional activity are unknown. METHODS Direct reprogramming of mouse fibroblasts to induced renal tubular epithelial cells was conducted both with wild-type HNF1B and with patient mutations. HNF1B was expressed in Xenopus ectodermal explants. Transcriptomic analysis by bulk RNA-Seq identified conserved targets with differentially regulated expression by the wild-type or R295C mutant. CRISPR/Cas9 genome editing in Xenopus embryos evaluated transcriptional targets in vivo . RESULTS HNF1B is essential for reprogramming mouse fibroblasts to induced renal tubular epithelial cells and induces development of ectopic renal organoids from pluripotent Xenopus cells. The mutation R295C retains reprogramming and inductive capacity but alters the expression of specific sets of downstream target genes instead of diminishing overall transcriptional activity of HNF1B. Surprisingly, targets associated with polycystic kidney disease were less affected than genes affected in congenital renal anomalies. Cross-species-conserved transcriptional targets were dysregulated in hnf1b CRISPR-depleted Xenopus embryos, confirming their dependence on hnf1b . CONCLUSIONS HNF1B activates an evolutionarily conserved program of target genes that disease-causing mutations selectively disrupt. These findings provide insights into the renal transcriptional network that controls nephrogenesis.
Collapse
Affiliation(s)
- Kelli Grand
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Martine Stoltz
- The University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Ludovica Rizzo
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Ruth Röck
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Michael M. Kaminski
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Nephrology and Medical Intensive Care, Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | | | - Maike Getwan
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Thomas Naert
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Roman Pichler
- The University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Soeren S. Lienkamp
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
- The University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
5
|
Veljačić Visković D, Lozić M, Vukoja M, Šoljić V, Vukojević K, Glavina Durdov M, Filipović N, Lozić B. Spatio-Temporal Expression Pattern of CAKUT Candidate Genes DLG1 and KIF12 during Human Kidney Development. Biomolecules 2023; 13:biom13020340. [PMID: 36830709 PMCID: PMC9953652 DOI: 10.3390/biom13020340] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023] Open
Abstract
We aimed to investigate expression of the novel susceptibility genes for CAKUT, DLG1 and KIF12, proposed by a systematic in silico approach, in developing and postnatal healthy human kidneys to provide information about their spatiotemporal expression pattern. We analyzed expression of their protein products by immunohistochemistry and immunofluorescence and quantified relative mRNA levels by RT-qPCR. Statistically significant differences in expression patterns were observed between certain developmental stages. Strong expression of DLG1 was observed in the developing kidney, with a gradual decrease from the first phase of kidney development (Ph1) until the third phase (Ph3), when most nephrons are formed; at later stages, the highest expression was observed in the tubules. KIF12 was highly expressed in the developing structures, especially in Ph1, with a gradual decrease until the postnatal phase, which would indicate a significant role in nephrogenesis. Co-localization of DLG1 and KIF12 was pronounced in Ph1, especially on the apical side of the tubular epithelial cells. Thereafter, their expression gradually became weaker and was only visible as punctate staining in Ph4. The direct association of DLG1 with KIF12 as control genes of normal kidney development may reveal their new functional aspect in renal tubular epithelial cells.
Collapse
Affiliation(s)
| | - Mirela Lozić
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21 000 Split, Croatia
- Correspondence: ; Tel.: +385-21-557-800
| | - Martina Vukoja
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
| | - Violeta Šoljić
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Faculty of Health Studies, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
| | - Katarina Vukojević
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21 000 Split, Croatia
- Laboratory of Morphology, Department of Histology and Embryology, School of Medicine, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Faculty of Health Studies, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Department of Anatomy, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, University of Split School of Medicine, 21 000 Split, Croatia
| | - Merica Glavina Durdov
- Department of Pathology, University Hospital Split, 21 000 Split, Croatia
- School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia
| | - Natalija Filipović
- Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Šoltanska 2, 21 000 Split, Croatia
- Department of Anatomy, University of Mostar, 88 000 Mostar, Bosnia and Herzegovina
- Center for Translational Research in Biomedicine, University of Split School of Medicine, 21 000 Split, Croatia
| | - Bernarda Lozić
- Paediatric Diseases Department, University Hospital of Split, Spinčićeva 1, 21 000 Split, Croatia
- School of Medicine, University of Split, Šoltanska 2, 21 000 Split, Croatia
| |
Collapse
|
6
|
Boettcher S, Simons M. Model organisms for functional validation in genetic renal disease. MED GENET-BERLIN 2022; 34:287-296. [PMID: 38836086 PMCID: PMC11006349 DOI: 10.1515/medgen-2022-2162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Functional validation is key for establishing new disease genes in human genetics. Over the years, model organisms have been utilized in a very effective manner to prove causality of genes or genetic variants for a wide variety of diseases. Also in hereditary renal disease, model organisms are very helpful for functional validation of candidate genes and variants identified by next-generation sequencing strategies and for obtaining insights into the pathophysiology. Due to high genetic conservation as well as high anatomical and physiological similarities with the human kidney, almost all genetic kidney diseases can be studied in the mouse. However, mouse work is time consuming and expensive, so there is a need for alternative models. In this review, we will provide an overview of model organisms used in renal research, focusing on mouse, zebrafish, frog, and fruit flies.
Collapse
Affiliation(s)
- Susanne Boettcher
- Sektion Nephrogenetik, Institute of Human Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| | - Matias Simons
- Sektion Nephrogenetik, Institute of Human Genetics, University Hospital Heidelberg, 69120 Heidelberg, Germany
| |
Collapse
|
7
|
Yasunaga T, Wiegel J, Bergen MD, Helmstädter M, Epting D, Paolini A, Çiçek Ö, Radziwill G, Engel C, Brox T, Ronneberger O, Walentek P, Ulbrich MH, Walz G. Microridge-like structures anchor motile cilia. Nat Commun 2022; 13:2056. [PMID: 35440631 PMCID: PMC9018822 DOI: 10.1038/s41467-022-29741-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 03/30/2022] [Indexed: 01/11/2023] Open
Abstract
Several tissues contain cells with multiple motile cilia that generate a fluid or particle flow to support development and organ functions; defective motility causes human disease. Developmental cues orient motile cilia, but how cilia are locked into their final position to maintain a directional flow is not understood. Here we find that the actin cytoskeleton is highly dynamic during early development of multiciliated cells (MCCs). While apical actin bundles become increasingly more static, subapical actin filaments are nucleated from the distal tip of ciliary rootlets. Anchorage of these subapical actin filaments requires the presence of microridge-like structures formed during MCC development, and the activity of Nonmuscle Myosin II. Optogenetic manipulation of Ezrin, a core component of the microridge actin-anchoring complex, or inhibition of Myosin Light Chain Kinase interfere with rootlet anchorage and orientation. These observations identify microridge-like structures as an essential component of basal body rootlet anchoring in MCCs.
Collapse
Affiliation(s)
- Takayuki Yasunaga
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Johannes Wiegel
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Max D Bergen
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Martin Helmstädter
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Daniel Epting
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Andrea Paolini
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schaenzlestrasse 1, 79104, Freiburg, Germany
| | - Özgün Çiçek
- Pattern Recognition and Image Processing, Department of Computer Science, University of Freiburg, Georges-Köhler-Allee 52, 79110, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Gerald Radziwill
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Christina Engel
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
| | - Thomas Brox
- Pattern Recognition and Image Processing, Department of Computer Science, University of Freiburg, Georges-Köhler-Allee 52, 79110, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Olaf Ronneberger
- Pattern Recognition and Image Processing, Department of Computer Science, University of Freiburg, Georges-Köhler-Allee 52, 79110, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Peter Walentek
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Maximilian H Ulbrich
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany
| | - Gerd Walz
- Department of Medicine IV, University Freiburg Medical Center, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, 79106, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany.
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, Schänzlestrasse 18, 79104, Freiburg, Germany.
| |
Collapse
|
8
|
Rocha C, Prinos P. Post-transcriptional and Post-translational Modifications of Primary Cilia: How to Fine Tune Your Neuronal Antenna. Front Cell Neurosci 2022; 16:809917. [PMID: 35295905 PMCID: PMC8918543 DOI: 10.3389/fncel.2022.809917] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 01/19/2022] [Indexed: 12/27/2022] Open
Abstract
Primary cilia direct cellular signaling events during brain development and neuronal differentiation. The primary cilium is a dynamic organelle formed in a multistep process termed ciliogenesis that is tightly coordinated with the cell cycle. Genetic alterations, such as ciliary gene mutations, and epigenetic alterations, such as post-translational modifications and RNA processing of cilia related factors, give rise to human neuronal disorders and brain tumors such as glioblastoma and medulloblastoma. This review discusses the important role of genetics/epigenetics, as well as RNA processing and post-translational modifications in primary cilia function during brain development and cancer formation. We summarize mouse and human studies of ciliogenesis and primary cilia activity in the brain, and detail how cilia maintain neuronal progenitor populations and coordinate neuronal differentiation during development, as well as how cilia control different signaling pathways such as WNT, Sonic Hedgehog (SHH) and PDGF that are critical for neurogenesis. Moreover, we describe how post-translational modifications alter cilia formation and activity during development and carcinogenesis, and the impact of missplicing of ciliary genes leading to ciliopathies and cell cycle alterations. Finally, cilia genetic and epigenetic studies bring to light cellular and molecular mechanisms that underlie neurodevelopmental disorders and brain tumors.
Collapse
Affiliation(s)
- Cecilia Rocha
- Early Drug Discovery Unit, Montreal Neurological Institute-Hospital, McGill University, Montreal, QC, Canada
- *Correspondence: Cecilia Rocha,
| | - Panagiotis Prinos
- Structural Genomics Consortium, University of Toronto, Toronto, ON, Canada
- Panagiotis Prinos,
| |
Collapse
|
9
|
Expression Pattern of α-Tubulin, Inversin and Its Target Dishevelled-1 and Morphology of Primary Cilia in Normal Human Kidney Development and Diseases. Int J Mol Sci 2021; 22:ijms22073500. [PMID: 33800671 PMCID: PMC8037028 DOI: 10.3390/ijms22073500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 03/26/2021] [Indexed: 12/11/2022] Open
Abstract
The spatiotemporal expression of α-tubulin, inversin and dishevelled-1 (DVL-1) proteins associated with the Wnt-signaling pathway, and primary cilia morphology were analyzed in developing kidneys (14th–38th developmental weeks), healthy postnatal (1.5- and 7-years old) and pathologically changed human kidneys, including multicystic dysplastic kidneys (MCDK), focal segmental glomerulosclerosis (FSGS) and nephrotic syndrome of the Finnish type (CNF). The analysis was performed by double immunofluorescence, electron microscopy, semiquantitative and statistical methods. Cytoplasmic co-expression of α-tubulin, inversin and DVL-1 was observed in the proximal convoluted tubules (pct), distal convoluted tubules (dct) and glomeruli (g) of analyzed tissues. During kidney development, the overall expression of α-tubulin, inversin and DVL-1 decreased, while in the postnatal period slightly increased. The highest expressions of α-tubulin and inversin characterized dct and g, while high DVL-1 characterized pct. α-tubulin, inversin and DVL-1 expression pattern in MCDK, FSGS and CNF kidneys significantly differed from the healthy control. Compared to healthy kidneys, pathologically changed kidneys had dysmorphic primary cilia. Different expression dynamics of α-tubulin, inversin and DVL-1 during kidney development could indicate that switch between the canonical and noncanonical Wnt-signaling is essential for normal kidney morphogenesis. In contrast, their disturbed expression in pathological kidneys might be associated with abnormal primary cilia, leading to chronic kidney diseases.
Collapse
|
10
|
Nephronophthisis gene products display RNA-binding properties and are recruited to stress granules. Sci Rep 2020; 10:15954. [PMID: 32994509 PMCID: PMC7524721 DOI: 10.1038/s41598-020-72905-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 05/05/2020] [Indexed: 12/15/2022] Open
Abstract
Mutations of cilia-associated molecules cause multiple developmental defects that are collectively termed ciliopathies. However, several ciliary proteins, involved in gating access to the cilium, also assume localizations at other cellular sites including the nucleus, where they participate in DNA damage responses to maintain tissue integrity. Molecular insight into how these molecules execute such diverse functions remains limited. A mass spectrometry screen for ANKS6-interacting proteins suggested an involvement of ANKS6 in RNA processing and/or binding. Comparing the RNA-binding properties of the known RNA-binding protein BICC1 with the three ankyrin-repeat proteins ANKS3, ANKS6 (NPHP16) and INVERSIN (NPHP2) confirmed that certain nephronophthisis (NPH) family members can interact with RNA molecules. We also observed that BICC1 and INVERSIN associate with stress granules in response to translational inhibition. Furthermore, BICC1 recruits ANKS3 and ANKS6 into TIA-1-positive stress granules after exposure to hippuristanol. Our findings uncover a novel function of NPH family members, and provide further evidence that NPH family members together with BICC1 are involved in stress responses to maintain tissue and organ integrity.
Collapse
|
11
|
The Role of Wnt Signalling in Chronic Kidney Disease (CKD). Genes (Basel) 2020; 11:genes11050496. [PMID: 32365994 PMCID: PMC7290783 DOI: 10.3390/genes11050496] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/25/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022] Open
Abstract
Chronic kidney disease (CKD) encompasses a group of diverse diseases that are associated with accumulating kidney damage and a decline in glomerular filtration rate (GFR). These conditions can be of an acquired or genetic nature and, in many cases, interactions between genetics and the environment also play a role in disease manifestation and severity. In this review, we focus on genetically inherited chronic kidney diseases and dissect the links between canonical and non-canonical Wnt signalling, and this umbrella of conditions that result in kidney damage. Most of the current evidence on the role of Wnt signalling in CKD is gathered from studies in polycystic kidney disease (PKD) and nephronophthisis (NPHP) and reveals the involvement of β-catenin. Nevertheless, recent findings have also linked planar cell polarity (PCP) signalling to CKD, with further studies being required to fully understand the links and molecular mechanisms.
Collapse
|
12
|
Bennett HW, Gustavsson AK, Bayas CA, Petrov PN, Mooney N, Moerner WE, Jackson PK. Novel fibrillar structure in the inversin compartment of primary cilia revealed by 3D single-molecule superresolution microscopy. Mol Biol Cell 2020; 31:619-639. [PMID: 31895004 PMCID: PMC7202064 DOI: 10.1091/mbc.e19-09-0499] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Primary cilia in many cell types contain a periaxonemal subcompartment called the inversin compartment. Four proteins have been found to assemble within the inversin compartment: INVS, ANKS6, NEK8, and NPHP3. The function of the inversin compartment is unknown, but it appears to be critical for normal development, including left–right asymmetry and renal tissue homeostasis. Here we combine superresolution imaging of human RPE1 cells, a classic model for studying primary cilia in vitro, with a genetic dissection of the protein–protein binding relationships that organize compartment assembly to develop a new structural model. We observe that INVS is the core structural determinant of a compartment composed of novel fibril-like substructures, which we identify here by three-dimensional single-molecule superresolution imaging. We find that NEK8 and ANKS6 depend on INVS for localization to these fibrillar assemblies and that ANKS6-NEK8 density within the compartment is regulated by NEK8. Together, NEK8 and ANKS6 are required downstream of INVS to localize and concentrate NPHP3 within the compartment. In the absence of these upstream components, NPHP3 is redistributed within cilia. These results provide a more detailed structure for the inversin compartment and introduce a new example of a membraneless compartment organized by protein–protein interactions.
Collapse
Affiliation(s)
- Henrietta W Bennett
- Baxter Laboratory, Department of Microbiology and Immunology and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Anna-Karin Gustavsson
- Department of Chemistry, Stanford University, Stanford, CA 94305.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm SE 17177, Sweden
| | - Camille A Bayas
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Petar N Petrov
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Nancie Mooney
- Baxter Laboratory, Department of Microbiology and Immunology and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - W E Moerner
- Department of Chemistry, Stanford University, Stanford, CA 94305
| | - Peter K Jackson
- Baxter Laboratory, Department of Microbiology and Immunology and Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
13
|
Abstract
Primary cilia project in a single copy from the surface of most vertebrate cell types; they detect and transmit extracellular cues to regulate diverse cellular processes during development and to maintain tissue homeostasis. The sensory capacity of primary cilia relies on the coordinated trafficking and temporal localization of specific receptors and associated signal transduction modules in the cilium. The canonical Hedgehog (HH) pathway, for example, is a bona fide ciliary signalling system that regulates cell fate and self-renewal in development and tissue homeostasis. Specific receptors and associated signal transduction proteins can also localize to primary cilia in a cell type-dependent manner; available evidence suggests that the ciliary constellation of these proteins can temporally change to allow the cell to adapt to specific developmental and homeostatic cues. Consistent with important roles for primary cilia in signalling, mutations that lead to their dysfunction underlie a pleiotropic group of diseases and syndromic disorders termed ciliopathies, which affect many different tissues and organs of the body. In this Review, we highlight central mechanisms by which primary cilia coordinate HH, G protein-coupled receptor, WNT, receptor tyrosine kinase and transforming growth factor-β (TGFβ)/bone morphogenetic protein (BMP) signalling and illustrate how defects in the balanced output of ciliary signalling events are coupled to developmental disorders and disease progression.
Collapse
|
14
|
Blackburn ATM, Miller RK. Modeling congenital kidney diseases in Xenopus laevis. Dis Model Mech 2019; 12:12/4/dmm038604. [PMID: 30967415 PMCID: PMC6505484 DOI: 10.1242/dmm.038604] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) occur in ∼1/500 live births and are a leading cause of pediatric kidney failure. With an average wait time of 3-5 years for a kidney transplant, the need is high for the development of new strategies aimed at reducing the incidence of CAKUT and preserving renal function. Next-generation sequencing has uncovered a significant number of putative causal genes, but a simple and efficient model system to examine the function of CAKUT genes is needed. Xenopus laevis (frog) embryos are well-suited to model congenital kidney diseases and to explore the mechanisms that cause these developmental defects. Xenopus has many advantages for studying the kidney: the embryos develop externally and are easily manipulated with microinjections, they have a functional kidney in ∼2 days, and 79% of identified human disease genes have a verified ortholog in Xenopus. This facilitates high-throughput screening of candidate CAKUT-causing genes. In this Review, we present the similarities between Xenopus and mammalian kidneys, highlight studies of CAKUT-causing genes in Xenopus and describe how common kidney diseases have been modeled successfully in this model organism. Additionally, we discuss several molecular pathways associated with kidney disease that have been studied in Xenopus and demonstrate why it is a useful model for studying human kidney diseases. Summary: Understanding how congenital kidney diseases arise is imperative to their treatment. Using Xenopus as a model will aid in elucidating kidney development and congenital kidney diseases.
Collapse
Affiliation(s)
- Alexandria T M Blackburn
- Pediatric Research Center, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX 77030, USA.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics and Epigenetics, Houston, TX 77030, USA
| | - Rachel K Miller
- Pediatric Research Center, Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center, Houston, TX 77030, USA .,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Genetics and Epigenetics, Houston, TX 77030, USA.,The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Program in Biochemistry and Cell Biology Houston, Houston, TX 77030, USA.,Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
15
|
Hoff S, Epting D, Falk N, Schroda S, Braun DA, Halbritter J, Hildebrandt F, Kramer-Zucker A, Bergmann C, Walz G, Lienkamp SS. The nucleoside-diphosphate kinase NME3 associates with nephronophthisis proteins and is required for ciliary function during renal development. J Biol Chem 2018; 293:15243-15255. [PMID: 30111592 PMCID: PMC6166740 DOI: 10.1074/jbc.ra117.000847] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Revised: 08/07/2018] [Indexed: 01/12/2023] Open
Abstract
Nephronophthisis (NPH) is an autosomal recessive renal disease leading to kidney failure in children and young adults. The protein products of the corresponding genes (NPHPs) are localized in primary cilia or their appendages. Only about 70% of affected individuals have a mutation in one of 100 renal ciliopathy genes, and no unifying pathogenic mechanism has been identified. Recently, some NPHPs, including NIMA-related kinase 8 (NEK8) and centrosomal protein 164 (CEP164), have been found to act in the DNA-damage response pathway and to contribute to genome stability. Here, we show that NME/NM23 nucleoside-diphosphate kinase 3 (NME3) that has recently been found to facilitate DNA-repair mechanisms binds to several NPHPs, including NEK8, CEP164, and ankyrin repeat and sterile α motif domain-containing 6 (ANKS6). Depletion of nme3 in zebrafish and Xenopus resulted in typical ciliopathy-associated phenotypes, such as renal malformations and left-right asymmetry defects. We further found that endogenous NME3 localizes to the basal body and that it associates also with centrosomal proteins, such as NEK6, which regulates cell cycle arrest after DNA damage. The ciliopathy-typical manifestations of NME3 depletion in two vertebrate in vivo models, the biochemical association of NME3 with validated NPHPs, and its localization to the basal body reveal a role for NME3 in ciliary function. We conclude that mutations in the NME3 gene may aggravate the ciliopathy phenotypes observed in humans.
Collapse
Affiliation(s)
- Sylvia Hoff
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daniel Epting
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Nathalie Falk
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Sophie Schroda
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Daniela A Braun
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Jan Halbritter
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Friedhelm Hildebrandt
- Department of Medicine, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts 02115
| | - Albrecht Kramer-Zucker
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Carsten Bergmann
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Human Genetics, Bioscientia, 55218 Ingelheim, Germany, and
| | - Gerd Walz
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
- Center for Biological Signaling Studies (BIOSS), 79104 Freiburg, Germany
| | - Soeren S Lienkamp
- From the Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany,
- Center for Biological Signaling Studies (BIOSS), 79104 Freiburg, Germany
| |
Collapse
|
16
|
Kulkarni NH, Smith RC, Blazer-Yost BL. Loss of inversin decreases transepithelial sodium transport in murine renal cells. Am J Physiol Cell Physiol 2017; 313:C664-C673. [PMID: 28978526 DOI: 10.1152/ajpcell.00359.2016] [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] [Indexed: 11/22/2022]
Abstract
Type II nephronophthisis (NPHP2) is an autosomal recessive renal cystic disorder characterized by mutations in the inversin gene. Humans and mice with mutations in inversin have enlarged cystic kidneys that may be due to fluid accumulation resulting from altered ion transport. To address this, transepithelial ion transport was measured in shRNA-mediated inversin-depleted mouse cortical collecting duct (mCCD) cells. Loss of inversin decreased the basal ion flux in mCCD cells compared with controls. Depletion of inversin decreased vasopressin-induced Na+ absorption but did not alter Cl- secretion by mCCD cells. Addition of amiloride, a specific blocker of the epithelial sodium channel (ENaC), abolished basal ion transport in both inversin knockdown and control cells, indicating ENaC involvement. Transcript levels of ENaC β-subunit were reduced in inversin-knockdown cells consistent with decreased ENaC activity. Furthermore, Nedd4l (neural precursor cell expressed, developmentally downregulated 4 like), an upstream negative regulator of ENaC, was evaluated. The relative amount of the phosphorylated, inactive Nedd4l was decreased in inversin-depleted cells consistent with decreased ENaC activity. The protein levels of Sgk1 (serum and glucocorticoid-inducible kinase), which phosphorylates Nedd4l, remained unchanged although the transcript levels were increased in inversin-depleted cells. Interestingly, mRNA and protein levels of Crtc2 (Creb-regulated transcription coactivator) kinase, a positive regulator of Sgk1, were decreased in inversin-depleted cells. Together these results suggest that loss of inversin decreases Na+ transport via ENaC, mediated in part by transcriptional and posttranslational regulation of Crtc2/Sgk1/Nedd4l axis as a contributory mechanism for enlarged kidneys in NPHP2.
Collapse
Affiliation(s)
- Nalini H Kulkarni
- Department of Biology, Indiana University-Purdue University at Indianapolis , Indianapolis, Indiana
| | - Rosamund C Smith
- Department of Biology, Indiana University-Purdue University at Indianapolis , Indianapolis, Indiana
| | - Bonnie L Blazer-Yost
- Department of Biology, Indiana University-Purdue University at Indianapolis , Indianapolis, Indiana
| |
Collapse
|
17
|
Novel biomarkers in kidney disease: roles for cilia, Wnt signalling and ATMIN in polycystic kidney disease. Biochem Soc Trans 2017; 44:1745-1751. [PMID: 27913685 DOI: 10.1042/bst20160124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Revised: 08/30/2016] [Accepted: 09/02/2016] [Indexed: 01/24/2023]
Abstract
Biomarkers, the measurable indicators of biological conditions, are fast becoming a popular approach in providing information to track disease processes that could lead to novel therapeutic interventions for chronic conditions. Inherited, chronic kidney disease affects millions of people worldwide and although pharmacological treatments exist for some conditions, there are still patients whose only option is kidney dialysis and kidney transplantation. In the past 10 years, certain chronic kidney diseases have been reclassified as ciliopathies. Cilia in the kidney are antenna-like, sensory organelles that are required for signal transduction. One of the signalling pathways that requires the primary cilium in the kidney is Wnt signalling and it has three components such as canonical Wnt, non-canonical Wnt/planar cell olarity (PCP) and non-canonical Wnt/Ca2+ signalling. Identification of the novel role of ATM INteractor (ATMIN) as an effector molecule in the non-canonical Wnt/PCP pathway has intrigued us to investigate its potential role in chronic kidney disease. ATMIN could thus be an important biomarker in disease prognosis and treatment that might lighten the burden of chronic kidney disease and also affect on its progression.
Collapse
|
18
|
Jiang GY, Zhang Y, Zhang XP, Lin XY, Yu JH, Wang EH. Inversin correlates with the malignant phenotype of non-small cell lung cancer and promotes the invasiveness of lung cancer cells. Tumour Biol 2017; 39:1010428317691177. [PMID: 28618971 DOI: 10.1177/1010428317691177] [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] [Indexed: 11/16/2022] Open
Abstract
Inversin, encoded by NPHP2, is one of the 10 NPHP proteins known to be involved in nephronophthisis (an autosomal recessive cystic kidney). Although the previous reports showed that inversin played an important role in embryonic development and renal diseases, its function in cancer was not revealed clearly so far. As measured by immunohistochemical staining, inversin was highly expressed in the cytoplasm of lung cancer samples (63.4%, 161/254) compared with adjacent normal lung tissues (22.0%, 11/50, p < 0.01). Moreover, its expression was positively correlated with differentiation ( p = 0.014), tumor node metastasis staging ( p = 0.007), and lymph node metastasis ( p = 0.020). The overall survival of non-small cell lung cancer patients with inversin positive expression (45.41 ± 1.800 months) was significantly reduced compared with those with inversin negative expression (51.046 ± 2.238 months, p = 0.042). Consistently, we found that the invasion capacity of A549 cells transfected with inversin was significantly stronger than that of control cells ( p < 0.05), while inversin siRNA-treatment significantly reduced cell invasion in H1299 cells ( p < 0.05). Additionally, we demonstrated that inversin could upregulate the expression of N-cadherin, Vimentin, matrix metalloproteinase-2, and matrix metalloproteinase-9. Collectively, these results indicated that inversin might promote the tumorigenicity of lung cancer cells and serve as a novel therapeutic target of non-small cell lung cancer.
Collapse
Affiliation(s)
- Gui-Yang Jiang
- 1 Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yong Zhang
- 2 Department of Pathology, Affiliated Tumor Hospital of China Medical University, Shenyang, China
| | - Xiu-Peng Zhang
- 1 Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xu-Yong Lin
- 1 Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Juan-Han Yu
- 1 Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - En-Hua Wang
- 1 Department of Pathology, First Affiliated Hospital and College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
19
|
Toolbox in a tadpole: Xenopus for kidney research. Cell Tissue Res 2017; 369:143-157. [PMID: 28401306 DOI: 10.1007/s00441-017-2611-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 03/09/2017] [Indexed: 01/14/2023]
Abstract
Xenopus is a versatile model organism increasingly used to study organogenesis and genetic diseases. The rapid embryonic development, targeted injections, loss- and gain-of-function experiments and an increasing supply of tools for functional in vivo analysis are unique advantages of the Xenopus system. Here, we review the vast array of methods available that have facilitated its transition into a translational model. We will focus primarily on how these methods have been employed in the study of kidney development, renal function and kidney disease. Future advances in the fields of genome editing, imaging and quantitative 'omics approaches are likely to enable exciting and novel applications for Xenopus to deepen our understanding of core principles of renal development and molecular mechanisms of human kidney disease. Thus, using Xenopus in clinically relevant research diversifies the narrowing pool of "standard" model organisms and provides unique opportunities for translational research.
Collapse
|
20
|
Vivante A, Mann N, Yonath H, Weiss AC, Getwan M, Kaminski MM, Bohnenpoll T, Teyssier C, Chen J, Shril S, van der Ven AT, Ityel H, Schmidt JM, Widmeier E, Bauer SB, Sanna-Cherchi S, Gharavi AG, Lu W, Magen D, Shukrun R, Lifton RP, Tasic V, Stanescu HC, Cavaillès V, Kleta R, Anikster Y, Dekel B, Kispert A, Lienkamp SS, Hildebrandt F. A Dominant Mutation in Nuclear Receptor Interacting Protein 1 Causes Urinary Tract Malformations via Dysregulation of Retinoic Acid Signaling. J Am Soc Nephrol 2017; 28:2364-2376. [PMID: 28381549 DOI: 10.1681/asn.2016060694] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 02/20/2017] [Indexed: 12/31/2022] Open
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of CKD in the first three decades of life. However, for most patients with CAKUT, the causative mutation remains unknown. We identified a kindred with an autosomal dominant form of CAKUT. By whole-exome sequencing, we identified a heterozygous truncating mutation (c.279delG, p.Trp93fs*) of the nuclear receptor interacting protein 1 gene (NRIP1) in all seven affected members. NRIP1 encodes a nuclear receptor transcriptional cofactor that directly interacts with the retinoic acid receptors (RARs) to modulate retinoic acid transcriptional activity. Unlike wild-type NRIP1, the altered NRIP1 protein did not translocate to the nucleus, did not interact with RARα, and failed to inhibit retinoic acid-dependent transcriptional activity upon expression in HEK293 cells. Notably, we also showed that treatment with retinoic acid enhanced NRIP1 binding to RARα RNA in situ hybridization confirmed Nrip1 expression in the developing urogenital system of the mouse. In explant cultures of embryonic kidney rudiments, retinoic acid stimulated Nrip1 expression, whereas a pan-RAR antagonist strongly reduced it. Furthermore, mice heterozygous for a null allele of Nrip1 showed a CAKUT-spectrum phenotype. Finally, expression and knockdown experiments in Xenopus laevis confirmed an evolutionarily conserved role for NRIP1 in renal development. These data indicate that dominant NRIP1 mutations can cause CAKUT by interference with retinoic acid transcriptional signaling, shedding light on the well documented association between abnormal vitamin A levels and renal malformations in humans, and suggest a possible gene-environment pathomechanism in this disease.
Collapse
Affiliation(s)
- Asaf Vivante
- Departments of Medicine and.,Talpiot Medical Leadership Program, Sheba Medical Center, Tel-Hashomer, Israel
| | | | - Hagith Yonath
- Department of Internal Medicine A and Genetics Institute, Sheba Medical Center and Sackler Faculty of Medicine Tel Aviv University, Tel Aviv, Israel
| | - Anna-Carina Weiss
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Maike Getwan
- Department of Medicine, Renal Division, University Medical Center, Faculty of Medicine, and
| | - Michael M Kaminski
- Department of Medicine, Renal Division, University Medical Center, Faculty of Medicine, and
| | - Tobias Bohnenpoll
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Catherine Teyssier
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Montpellier, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France; Université Montpellier, Montpellier, France; Institut régional du Cancer de Montpellier, Montpellier, France
| | | | | | | | | | | | - Eugen Widmeier
- Departments of Medicine and.,Department of Medicine, Renal Division, University Medical Center, Faculty of Medicine, and
| | - Stuart B Bauer
- Urology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Ali G Gharavi
- Division of Nephrology, Columbia University, New York, New York
| | - Weining Lu
- Renal Section, Department of Medicine, Boston University Medical Center, Boston, Massachusetts
| | - Daniella Magen
- Pediatric Nephrology Institute, Rambam Health Care Campus, and Technion-Israel Institute of Technology, Haifa, Israel
| | - Rachel Shukrun
- Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Richard P Lifton
- Department of Human Genetics, Yale University School of Medicine, New Haven, Connecticut.,Howard Hughes Medical Institute, Chevy Chase, Maryland
| | - Velibor Tasic
- Department of Pediatric Nephrology, Medical Faculty Skopje, University Children's Hospital, Skopje, Macedonia; and
| | - Horia C Stanescu
- Centre for Nephrology, University College London, London, United Kingdom
| | - Vincent Cavaillès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), Montpellier, France; Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France; Université Montpellier, Montpellier, France; Institut régional du Cancer de Montpellier, Montpellier, France
| | - Robert Kleta
- Centre for Nephrology, University College London, London, United Kingdom
| | - Yair Anikster
- Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Benjamin Dekel
- Department of Pediatrics, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel-Hashomer, Israel and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Andreas Kispert
- Institut für Molekularbiologie, Medizinische Hochschule Hannover, Hannover, Germany
| | - Soeren S Lienkamp
- Department of Medicine, Renal Division, University Medical Center, Faculty of Medicine, and.,Center for Biological Signaling Studies (BIOSS), Albert Ludwig University, Freiburg, Germany
| | | |
Collapse
|
21
|
Krneta-Stankic V, DeLay BD, Miller RK. Xenopus: leaping forward in kidney organogenesis. Pediatr Nephrol 2017; 32:547-555. [PMID: 27099217 PMCID: PMC5074909 DOI: 10.1007/s00467-016-3372-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/08/2016] [Accepted: 03/09/2016] [Indexed: 12/17/2022]
Abstract
While kidney donations stagnate, the number of people in need of kidney transplants continues to grow. Although transplanting culture-grown organs is years away, pursuing the engineering of the kidney de novo is a valid means of closing the gap between the supply and demand of kidneys for transplantation. The structural organization of a mouse kidney is similar to that of humans. Therefore, mice have traditionally served as the primary model system for the study of kidney development. The mouse is an ideal model organism for understanding the complexity of the human kidney. Nonetheless, the elaborate structure of the mammalian kidney makes the discovery of new therapies based on de novo engineered kidneys more challenging. In contrast to mammals, amphibians have a kidney that is anatomically less complex and develops faster. Given that analogous genetic networks regulate the development of mammalian and amphibian nephric organs, using embryonic kidneys of Xenopus laevis (African clawed frog) to analyze inductive cell signaling events and morphogenesis has many advantages. Pioneering work that led to the ability to generate kidney organoids from embryonic cells was carried out in Xenopus. In this review, we discuss how Xenopus can be utilized to compliment the work performed in mammalian systems to understand kidney development.
Collapse
Affiliation(s)
- Vanja Krneta-Stankic
- Department of Pediatrics, Pediatric Research Center, University of Texas McGovern Medical School, 6431 Fannin Street, MSE R413, Houston, TX, 77030, USA
- Program in Genes and Development, University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Bridget D DeLay
- Department of Pediatrics, Pediatric Research Center, University of Texas McGovern Medical School, 6431 Fannin Street, MSE R413, Houston, TX, 77030, USA
| | - Rachel K Miller
- Department of Pediatrics, Pediatric Research Center, University of Texas McGovern Medical School, 6431 Fannin Street, MSE R413, Houston, TX, 77030, USA.
- Program in Genes and Development, University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA.
- Program in Cell and Regulatory Biology, University of Texas Graduate School of Biomedical Sciences, Houston, TX, USA.
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
22
|
Pronephric tubule formation in zebrafish: morphogenesis and migration. Pediatr Nephrol 2017; 32:211-216. [PMID: 26942753 DOI: 10.1007/s00467-016-3353-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 01/14/2023]
Abstract
The nephron is the functional subunit of the vertebrate kidney and plays important osmoregulatory and excretory roles during embryonic development and in adulthood. Despite its central role in kidney function, surprisingly little is known about the molecular and cellular processes that control nephrogenesis. The zebrafish pronephric kidney, comprising two nephrons, provides a visually accessible and genetically tractable model system for a better understanding of nephron formation. Using this system, various developmental processes, including the commitment of mesoderm to a kidney fate, renal tubule proliferation, and migration, can be studied during nephrogenesis. Here, we discuss some of these processes in zebrafish with a focus on the pathways that influence renal tubule cell morphogenesis.
Collapse
|
23
|
Walentek P, Quigley IK. What we can learn from a tadpole about ciliopathies and airway diseases: Using systems biology in Xenopus to study cilia and mucociliary epithelia. Genesis 2017; 55:10.1002/dvg.23001. [PMID: 28095645 PMCID: PMC5276738 DOI: 10.1002/dvg.23001] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 11/14/2016] [Accepted: 11/16/2016] [Indexed: 12/11/2022]
Abstract
Over the past years, the Xenopus embryo has emerged as an incredibly useful model organism for studying the formation and function of cilia and ciliated epithelia in vivo. This has led to a variety of findings elucidating the molecular mechanisms of ciliated cell specification, basal body biogenesis, cilia assembly, and ciliary motility. These findings also revealed the deep functional conservation of signaling, transcriptional, post-transcriptional, and protein networks employed in the formation and function of vertebrate ciliated cells. Therefore, Xenopus research can contribute crucial insights not only into developmental and cell biology, but also into the molecular mechanisms underlying cilia related diseases (ciliopathies) as well as diseases affecting the ciliated epithelium of the respiratory tract in humans (e.g., chronic lung diseases). Additionally, systems biology approaches including transcriptomics, genomics, and proteomics have been rapidly adapted for use in Xenopus, and broaden the applications for current and future translational biomedical research. This review aims to present the advantages of using Xenopus for cilia research, highlight some of the evolutionarily conserved key concepts and mechanisms of ciliated cell biology that were elucidated using the Xenopus model, and describe the potential for Xenopus research to address unresolved questions regarding the molecular mechanisms of ciliopathies and airway diseases.
Collapse
Affiliation(s)
- Peter Walentek
- Department of Molecular and Cell Biology; Genetics, Genomics and Development Division; Developmental and Regenerative Biology Group; University of California, Berkeley, CA 94720, USA
| | - Ian K. Quigley
- Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| |
Collapse
|
24
|
Kaminski MM, Tosic J, Kresbach C, Engel H, Klockenbusch J, Müller AL, Pichler R, Grahammer F, Kretz O, Huber TB, Walz G, Arnold SJ, Lienkamp SS. Direct reprogramming of fibroblasts into renal tubular epithelial cells by defined transcription factors. Nat Cell Biol 2016; 18:1269-1280. [PMID: 27820600 DOI: 10.1038/ncb3437] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/06/2016] [Indexed: 12/12/2022]
Abstract
Direct reprogramming by forced expression of transcription factors can convert one cell type into another. Thus, desired cell types can be generated bypassing pluripotency. However, direct reprogramming towards renal cells remains an unmet challenge. Here, we identify renal cell fate-inducing factors on the basis of their tissue specificity and evolutionarily conserved expression, and demonstrate that combined expression of Emx2, Hnf1b, Hnf4a and Pax8 converts mouse and human fibroblasts into induced renal tubular epithelial cells (iRECs). iRECs exhibit epithelial features, a global gene expression profile resembling their native counterparts, functional properties of differentiated renal tubule cells and sensitivity to nephrotoxic substances. Furthermore, iRECs integrate into kidney organoids and form tubules in decellularized kidneys. Our approach demonstrates that reprogramming factors can be identified by targeted in silico analysis. Renal tubular epithelial cells generated ex vivo by forced expression of transcription factors may facilitate disease modelling, drug and nephrotoxicity testing, and regenerative approaches.
Collapse
Affiliation(s)
- Michael M Kaminski
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Jelena Tosic
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Faculty of Medicine, Albertstraße 25, 79104 Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Albertstraße 19a, 79104 Freiburg, Germany.,Faculty of Biology, University of Freiburg, Schänzlestraße 1, 79104 Freiburg, Germany
| | - Catena Kresbach
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Hannes Engel
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Jonas Klockenbusch
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Anna-Lena Müller
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Roman Pichler
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Florian Grahammer
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany
| | - Oliver Kretz
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany.,Department of Neuroanatomy, University of Freiburg, Albertstraße 17, 79104 Freiburg, Germany
| | - Tobias B Huber
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany.,BIOSS Centre of Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Gerd Walz
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany.,BIOSS Centre of Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Sebastian J Arnold
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Faculty of Medicine, Albertstraße 25, 79104 Freiburg, Germany.,BIOSS Centre of Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| | - Soeren S Lienkamp
- Department of Medicine, Renal Division, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Straße 55, 79106 Freiburg, Germany.,BIOSS Centre of Biological Signalling Studies, University of Freiburg, Schänzlestraße 18, 79104 Freiburg, Germany
| |
Collapse
|
25
|
Yasunaga T, Hoff S, Schell C, Helmstädter M, Kretz O, Kuechlin S, Yakulov TA, Engel C, Müller B, Bensch R, Ronneberger O, Huber TB, Lienkamp SS, Walz G. The polarity protein Inturned links NPHP4 to Daam1 to control the subapical actin network in multiciliated cells. J Cell Biol 2016; 211:963-73. [PMID: 26644512 PMCID: PMC4674276 DOI: 10.1083/jcb.201502043] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Inturned-mediated complex formation of NPHP4 and DAAM1 is important for ciliogenesis and ciliary function in multiciliated cells, presumably because of its requirement for the local rearrangement of actin cytoskeleton. Motile cilia polarization requires intracellular anchorage to the cytoskeleton; however, the molecular machinery that supports this process remains elusive. We report that Inturned plays a central role in coordinating the interaction between cilia-associated proteins and actin-nucleation factors. We observed that knockdown of nphp4 in multiciliated cells of the Xenopus laevis epidermis compromised ciliogenesis and directional fluid flow. Depletion of nphp4 disrupted the subapical actin layer. Comparison to the structural defects caused by inturned depletion revealed striking similarities. Furthermore, coimmunoprecipitation assays demonstrated that the two proteins interact with each other and that Inturned mediates the formation of ternary protein complexes between NPHP4 and DAAM1. Knockdown of daam1, but not formin-2, resulted in similar disruption of the subapical actin web, whereas nphp4 depletion prevented the association of Inturned with the basal bodies. Thus, Inturned appears to function as an adaptor protein that couples cilia-associated molecules to actin-modifying proteins to rearrange the local actin cytoskeleton.
Collapse
Affiliation(s)
- Takayuki Yasunaga
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Sylvia Hoff
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Christoph Schell
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Martin Helmstädter
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Oliver Kretz
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Neuroanatomy, University of Freiburg, 79104 Freiburg, Germany
| | - Sebastian Kuechlin
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Toma A Yakulov
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Christina Engel
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Barbara Müller
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany
| | - Robert Bensch
- Department of Computer Science, University of Freiburg, 79110 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Olaf Ronneberger
- Department of Computer Science, University of Freiburg, 79110 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Tobias B Huber
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Soeren S Lienkamp
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Gerd Walz
- Renal Division, Department of Medicine, University of Freiburg Medical Center, 79106 Freiburg, Germany Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| |
Collapse
|
26
|
Lienkamp SS. Using Xenopus to study genetic kidney diseases. Semin Cell Dev Biol 2016; 51:117-24. [PMID: 26851624 DOI: 10.1016/j.semcdb.2016.02.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/01/2016] [Indexed: 10/22/2022]
Abstract
Modern sequencing technology is revolutionizing our knowledge of inherited kidney disease. However, the molecular role of genes affected by the rapidly rising number of identified mutations is lagging behind. Xenopus is a highly useful, but underutilized model organism with unique properties excellently suited to decipher the molecular mechanisms of kidney development and disease. The embryonic kidney (pronephros) can be manipulated on only one side of the animal and its formation observed directly through the translucent skin. The moderate evolutionary distance between Xenopus and humans is a huge advantage for studying basic principles of kidney development, but still allows us to analyze the function of disease related genes. Optogenetic manipulations and genome editing by CRISPR/Cas are exciting additions to the toolbox for disease modelling and will facilitate the use of Xenopus in translational research. Therefore, the future of Xenopus in kidney research is bright.
Collapse
Affiliation(s)
- Soeren S Lienkamp
- Renal Division, Department of Medicine, University of Freiburg Medical Center, Hugstetter Straße 55, 79106 Freiburg, Germany; Center for Biological Signaling Studies (BIOSS), Albertstraße 19, 79104 Freiburg, Germany.
| |
Collapse
|
27
|
3D U-Net: Learning Dense Volumetric Segmentation from Sparse Annotation. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION – MICCAI 2016 2016. [DOI: 10.1007/978-3-319-46723-8_49] [Citation(s) in RCA: 1382] [Impact Index Per Article: 153.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
28
|
Sun J, Wang X, Li C, Mao B. Xenopus Claudin-6 is required for embryonic pronephros morphogenesis and terminal differentiation. Biochem Biophys Res Commun 2015; 462:178-83. [DOI: 10.1016/j.bbrc.2015.04.065] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 04/11/2015] [Indexed: 11/16/2022]
|
29
|
Anks3 interacts with nephronophthisis proteins and is required for normal renal development. Kidney Int 2015; 87:1191-200. [DOI: 10.1038/ki.2015.17] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 11/10/2014] [Accepted: 12/05/2014] [Indexed: 12/19/2022]
|
30
|
Wang Y, Sun ZH, Zhou L, Li Z, Gui JF. Grouper tshβ promoter-driven transgenic zebrafish marks proximal kidney tubule development. PLoS One 2014; 9:e97806. [PMID: 24905828 PMCID: PMC4048157 DOI: 10.1371/journal.pone.0097806] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 04/24/2014] [Indexed: 01/09/2023] Open
Abstract
Kidney tubule plays a critical role in recovering or secreting solutes, but the detailed morphogenesis remains unclear. Our previous studies have found that grouper tshβ (gtshβ) is also expressed in kidney, however, the distribution significance is still unknown. To understand the gtshβ role and kidney tubule morphogenesis, here, we have generated a transgenic zebrafish line Tg(gtshβ:GFP) with green fluorescent protein driven by the gtshβ promoter. Similar to the endogenous tshβ in zebrafish or in grouper, the gtshβ promoter-driven GFP is expressed in pituitary and kidney, and the developing details of proximal kidney tubule are marked in the transgenic zebrafish line. The gfp initially transcribes at 16 hours post fertilization (hpf) above the dorsal mesentery, and partially co-localizes with pronephric tubular markers slc20a1a and cdh17. Significantly, the GFP specifically localizes in proximal pronephric segments during embryogenesis and resides at kidney duct epithelium in adult fish. To test whether the gtshβ promoter-driven GFP may serve as a readout signal of the tubular development, we have treated the embryos with retinoic acid signaing (RA) reagents, in which exogenous RA addition results in a distal extension of the proximal segments, while RA inhibition induces a weakness and shortness of the proximal segments. Therefore, this transgenic line provides a useful tool for genetic or chemical analysis of kidney tubule.
Collapse
Affiliation(s)
- Yang Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Wuhan, China
| | - Zhi-Hui Sun
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Wuhan, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Wuhan, China
| | - Zhi Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Wuhan, China
| | - Jian-Fang Gui
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, University of the Chinese Academy of Sciences, Wuhan, China
| |
Collapse
|
31
|
Cizelsky W, Tata A, Kühl M, Kühl SJ. The Wnt/JNK signaling target gene alcam is required for embryonic kidney development. Development 2014; 141:2064-74. [PMID: 24764076 DOI: 10.1242/dev.107938] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Proper development of nephrons is essential for kidney function. β-Catenin-independent Wnt signaling through Fzd8, Inversin, Daam1, RhoA and Myosin is required for nephric tubule morphogenesis. Here, we provide a novel mechanism through which non-canonical Wnt signaling contributes to tubular development. Using Xenopus laevis as a model system, we found that the cell-adhesion molecule Alcam is required for proper nephrogenesis and functions downstream of Fzd3 during embryonic kidney development. We found alcam expression to be independent of Fzd8 or Inversin, but to be transcriptionally regulated by the β-Catenin-independent Wnt/JNK pathway involving ATF2 and Pax2 in a direct manner. These novel findings indicate that several branches of Wnt signaling are independently required for proximal tubule development. Moreover, our data indicate that regulation of morphogenesis by non-canonical Wnt ligands also involves direct transcriptional responses in addition to the effects on a post-translational level.
Collapse
Affiliation(s)
- Wiebke Cizelsky
- Institute for Biochemistry and Molecular Biology, Ulm University, Albert-Einstein-Allee 11, Ulm 89081, Germany
| | | | | | | |
Collapse
|
32
|
Blacque OE, Sanders AAWM. Compartments within a compartment: what C. elegans can tell us about ciliary subdomain composition, biogenesis, function, and disease. Organogenesis 2014; 10:126-37. [PMID: 24732235 DOI: 10.4161/org.28830] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The primary cilium has emerged as a hotbed of sensory and developmental signaling, serving as a privileged domain to concentrate the functions of a wide number of channels, receptors and downstream signal transducers. This realization has provided important insight into the pathophysiological mechanisms underlying the ciliopathies, an ever expanding spectrum of multi-symptomatic disorders affecting the development and maintenance of multiple tissues and organs. One emerging research focus is the subcompartmentalised nature of the organelle, consisting of discrete structural and functional subdomains such as the periciliary membrane/basal body compartment, the transition zone, the Inv compartment and the distal segment/ciliary tip region. Numerous ciliopathy, transport-related and signaling molecules localize at these compartments, indicating specific roles at these subciliary sites. Here, by focusing predominantly on research from the genetically tractable nematode C. elegans, we review ciliary subcompartments in terms of their structure, function, composition, biogenesis and relationship to human disease.
Collapse
Affiliation(s)
- Oliver E Blacque
- School of Biomolecular and Biomedical Science; University College Dublin; Dublin, Ireland
| | - Anna A W M Sanders
- School of Biomolecular and Biomedical Science; University College Dublin; Dublin, Ireland
| |
Collapse
|
33
|
Abstract
Cystic kidney diseases can cause end stage renal disease, affecting millions of individuals worldwide. They may arise early or later in life, are characterized by a spectrum of symptoms and can be caused by diverse genetic defects. The primary cilium, a microtubule-based organelle that can serve as a signaling antenna, has been demonstrated to have a significant role in ensuring correct kidney development and function. In the kidney, one of the signaling pathways that requires the cilium for normal development is Wnt signaling. In this review, the roles of primary cilia in relation to canonical and non-canonical Wnt/PCP signaling in cystic renal disease are described. The evidence of the associations between cilia, Wnt signaling and cystic renal disease is discussed and the significance of planar cell polarity-related mechanisms in cystic kidney disease is presented. Although defective Wnt signaling is not the only cause of renal disease, research is increasingly highlighting its importance, encouraging the development of Wnt-associated diagnostic and prognostic tools for cystic renal disease.
Collapse
|
34
|
Fedeles S, Gallagher AR. Cell polarity and cystic kidney disease. Pediatr Nephrol 2013; 28:1161-72. [PMID: 23161205 DOI: 10.1007/s00467-012-2337-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/02/2012] [Accepted: 10/02/2012] [Indexed: 10/27/2022]
Abstract
Epithelial cell polarity is essential for organ development; aberrations in this process have been implicated in various diseases, including polycystic kidney disease. Establishment and maintenance of cell polarity is governed by a number of molecular processes and how these processes operate remains an interesting question. Conserved protein complexes guide both apical-basolateral polarity and planar cell polarity. In this review we discuss the recent findings that provide insights into polarity mechanisms and the intriguing crosstalk between apical-basolateral polarity and planar cell polarity, and their relationship to cystic kidney disease.
Collapse
Affiliation(s)
- Sorin Fedeles
- Section of Nephrology, Department of Internal Medicine, Yale University School of Medicine, P.O. Box 208029, 333 Cedar Street, New Haven, CT 06520-8029, USA
| | | |
Collapse
|
35
|
Hoff S, Halbritter J, Epting D, Frank V, Nguyen TMT, van Reeuwijk J, Boehlke C, Schell C, Yasunaga T, Helmstädter M, Mergen M, Filhol E, Boldt K, Horn N, Ueffing M, Otto EA, Eisenberger T, Elting MW, van Wijk JAE, Bockenhauer D, Sebire NJ, Rittig S, Vyberg M, Ring T, Pohl M, Pape L, Neuhaus TJ, Elshakhs NAS, Koon SJ, Harris PC, Grahammer F, Huber TB, Kuehn EW, Kramer-Zucker A, Bolz HJ, Roepman R, Saunier S, Walz G, Hildebrandt F, Bergmann C, Lienkamp SS. ANKS6 is a central component of a nephronophthisis module linking NEK8 to INVS and NPHP3. Nat Genet 2013; 45:951-6. [PMID: 23793029 PMCID: PMC3786259 DOI: 10.1038/ng.2681] [Citation(s) in RCA: 156] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 06/03/2013] [Indexed: 11/09/2022]
Abstract
Nephronophthisis (NPH) is an autosomal recessive cystic kidney disease that leads to renal failure in childhood or adolescence. Most NPHP gene products form molecular networks. We have identified ANKS6 as a new NPHP family member that connects NEK8 (NPHP9) to INVERSIN (INVS, NPHP2) and NPHP3 to form a distinct NPHP module. ANKS6 localizes to the proximal cilium and knockdown experiments in zebrafish and Xenopus confirmed a role in renal development. Genetic screening identified six families with ANKS6 mutations and NPH, including severe cardiovascular abnormalities, liver fibrosis and situs inversus. The oxygen sensor HIF1AN (FIH) hydroxylates ANKS6 and INVS, while knockdown of Hif1an in Xenopus resembled the loss of other NPHP proteins. HIF1AN altered the composition of the ANKS6/INVS/NPHP3 module. Network analyses, uncovering additional putative NPHP-associated genes, placed ANKS6 at the center of the NPHP module, explaining the overlapping disease manifestation caused by mutations of either ANKS6, NEK8, INVS or NPHP3.
Collapse
Affiliation(s)
- Sylvia Hoff
- Department of Medicine, Renal Division, University of Freiburg Medical Center, Freiburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Veland IR, Montjean R, Eley L, Pedersen LB, Schwab A, Goodship J, Kristiansen K, Pedersen SF, Saunier S, Christensen ST. Inversin/Nephrocystin-2 is required for fibroblast polarity and directional cell migration. PLoS One 2013; 8:e60193. [PMID: 23593172 PMCID: PMC3620528 DOI: 10.1371/journal.pone.0060193] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 02/22/2013] [Indexed: 12/30/2022] Open
Abstract
Inversin is a ciliary protein that critically regulates developmental processes and tissue homeostasis in vertebrates, partly through the degradation of Dishevelled (Dvl) proteins to coordinate Wnt signaling in planar cell polarity (PCP). Here, we investigated the role of Inversin in coordinating cell migration, which highly depends on polarity processes at the single-cell level, including the spatial and temporal organization of the cytoskeleton as well as expression and cellular localization of proteins in leading edge formation of migrating cells. Using cultures of mouse embryonic fibroblasts (MEFs) derived from inv(-/-) and inv(+/+) animals, we confirmed that both inv(-/-) and inv(+/+) MEFs form primary cilia, and that Inversin localizes to the primary cilium in inv(+/+) MEFs. In wound healing assays, inv(-/-) MEFs were severely compromised in their migratory ability and exhibited cytoskeletal rearrangements, including distorted lamellipodia formation and cilia orientation. Transcriptome analysis revealed dysregulation of Wnt signaling and of pathways regulating actin organization and focal adhesions in inv(-/-) MEFs as compared to inv(+/+) MEFs. Further, Dvl-1 and Dvl-3 localized to MEF primary cilia, and β-catenin/Wnt signaling was elevated in inv(-/-) MEFs, which moreover showed reduced ciliary localization of Dvl-3. Finally, inv(-/-) MEFs displayed dramatically altered activity and localization of RhoA, Rac1, and Cdc42 GTPases, and aberrant expression and targeting of the Na(+)/H(+) exchanger NHE1 and ezrin/radixin/moesin (ERM) proteins to the edge of cells facing the wound. Phosphorylation of β-catenin at the ciliary base and formation of well-defined lamellipodia with localization and activation of ERM to the leading edge of migrating cells were restored in inv(-/-) MEFs expressing Inv-GFP. Collectively, our findings point to the significance of Inversin in controlling cell migration processes, at least in part through transcriptional regulation of genes involved in Wnt signaling and pathways that control cytoskeletal organization and ion transport.
Collapse
Affiliation(s)
- Iben R. Veland
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Rodrick Montjean
- Inserm U-983, Imagine Institut, Paris Descartes-Sorbonne Paris Cité University, Necker Hospital, Paris, France
| | - Lorraine Eley
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Lotte B. Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Albrecht Schwab
- Institute of Physiology II, Münster University, Münster, Germany
| | - Judith Goodship
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, United Kingdom
| | | | - Stine F. Pedersen
- Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Saunier
- Inserm U-983, Imagine Institut, Paris Descartes-Sorbonne Paris Cité University, Necker Hospital, Paris, France
| | | |
Collapse
|
37
|
Zhang B, Romaker D, Ferrell N, Wessely O. Regulation of G-protein signaling via Gnas is required to regulate proximal tubular growth in the Xenopus pronephros. Dev Biol 2013; 376:31-42. [PMID: 23352791 DOI: 10.1016/j.ydbio.2013.01.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2012] [Revised: 01/10/2013] [Accepted: 01/14/2013] [Indexed: 11/17/2022]
Abstract
In the kidney, proximal tubules are very important for the reabsorption of water, ions and organic solutes from the primary urine. They are composed of highly specialized epithelial cells that are characterized by an elaborate apical brush border to increase transport efficiency. Using the pronephric kidney of Xenopus laevis we discovered that the G-protein modulator cholera toxin resulted in a dramatic reduction of the proximal tubular size. This phenotype was accompanied by changes in the cytoarchitecture characterized by ectopic expression of the distal tubular marker 4A6 and an impairment of yolk platelet degradation. In addition, cholera toxin caused edema formation. However, this phenotype was not due to kidney defects, but rather due to impaired vasculature development. Based on experiments with antisense morpholino oligomers as well as pharmacological agonists and antagonists, we could show that the complex phenotype of cholera toxin in the pronephric kidney was caused by the hyperactivation of a single G-protein alpha subunit, Gnas. This-in turn-caused elevated cAMP levels, triggered a Rapgef4-dependent signaling cassette and perturbed exo- and endocytosis. This perturbation of the secretory pathway by Ctx was not only observed in Xenopus embryos. Also, in a human proximal tubular cell line, cholera toxin or a Rapgef4-specific agonist increased uptake and decreased secretion of FITC-labeled Albumin. Based on these data we propose that the Gnas/cAMP/Rapgef4 pathway regulates the signals inducing the proliferation of proximal tubules to acquire their final organ size.
Collapse
Affiliation(s)
- Bo Zhang
- Cleveland Clinic Foundation, Lerner Research Institute, Department of Cellular and Molecular Medicine, 9500 Euclid Avenue/NC10, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
38
|
Vertebrate kidney tubules elongate using a planar cell polarity-dependent, rosette-based mechanism of convergent extension. Nat Genet 2012; 44:1382-7. [PMID: 23143599 DOI: 10.1038/ng.2452] [Citation(s) in RCA: 178] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 10/03/2012] [Indexed: 12/20/2022]
Abstract
Cystic kidney diseases are a global public health burden, affecting over 12 million people. Although much is known about the genetics of kidney development and disease, the cellular mechanisms driving normal kidney tubule elongation remain unclear. Here, we used in vivo imaging to show for the first time that mediolaterally oriented cell intercalation is fundamental to vertebrate kidney morphogenesis. Unexpectedly, we found that kidney tubule elongation is driven in large part by a myosin-dependent, multicellular rosette-based mechanism, previously only described in Drosophila melanogaster. In contrast to findings in Drosophila, however, non-canonical Wnt and planar cell polarity (PCP) signaling is required to control rosette topology and orientation during vertebrate kidney tubule elongation. These data resolve long-standing questions concerning the role of PCP signaling in the developing kidney and, moreover, establish rosette-based intercalation as a deeply conserved cellular engine for epithelial morphogenesis.
Collapse
|
39
|
McCooke JK, Appels R, Barrero RA, Ding A, Ozimek-Kulik JE, Bellgard MI, Morahan G, Phillips JK. A novel mutation causing nephronophthisis in the Lewis polycystic kidney rat localises to a conserved RCC1 domain in Nek8. BMC Genomics 2012; 13:393. [PMID: 22899815 PMCID: PMC3441220 DOI: 10.1186/1471-2164-13-393] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Accepted: 08/06/2012] [Indexed: 01/03/2023] Open
Abstract
Background Nephronophthisis (NPHP) as a cause of cystic kidney disease is the most common genetic cause of progressive renal failure in children and young adults. NPHP is characterized by abnormal and/or loss of function of proteins associated with primary cilia. Previously, we characterized an autosomal recessive phenotype of cystic kidney disease in the Lewis Polycystic Kidney (LPK) rat. Results In this study, quantitative trait locus analysis was used to define a ~1.6Mbp region on rat chromosome 10q25 harbouring the lpk mutation. Targeted genome capture and next-generation sequencing of this region identified a non-synonymous mutation R650C in the NIMA (never in mitosis gene a)- related kinase 8 ( Nek8) gene. This is a novel Nek8 mutation that occurs within the regulator of chromosome condensation 1 (RCC1)-like region of the protein. Specifically, the R650C substitution is located within a G[QRC]LG repeat motif of the predicted seven bladed beta-propeller structure of the RCC1 domain. The rat Nek8 gene is located in a region syntenic to portions of human chromosome 17 and mouse 11. Scanning electron microscopy confirmed abnormally long cilia on LPK kidney epithelial cells, and fluorescence immunohistochemistry for Nek8 protein revealed altered cilia localisation. Conclusions When assessed relative to other Nek8 NPHP mutations, our results indicate the whole propeller structure of the RCC1 domain is important, as the different mutations cause comparable phenotypes. This study establishes the LPK rat as a novel model system for NPHP and further consolidates the link between cystic kidney disease and cilia proteins.
Collapse
Affiliation(s)
- John K McCooke
- Centre for Comparative Genomics, Murdoch University, Perth, WA 6150, Australia
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Pan J, Seeger-Nukpezah T, Golemis EA. The role of the cilium in normal and abnormal cell cycles: emphasis on renal cystic pathologies. Cell Mol Life Sci 2012; 70:1849-74. [PMID: 22782110 DOI: 10.1007/s00018-012-1052-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Revised: 05/08/2012] [Accepted: 06/05/2012] [Indexed: 12/28/2022]
Abstract
The primary cilium protrudes from the cell surface and acts as a sensor for chemical and mechanical growth cues, with receptors for a number of growth factors (PDGFα, Hedgehog, Wnt, Notch) concentrated within the ciliary membrane. In normal tissues, the cilium assembles after cells exit mitosis and is resorbed as part of cell cycle re-entry. Although regulation of the cilium by cell cycle transitions has been appreciated for over 100 years, only recently have data emerged to indicate the cilium also exerts influence on the cell cycle. The resorption/protrusion cycle, regulated by proteins including Aurora-A, VHL, and GSK-3β, influences cell responsiveness to growth cues involving cilia-linked receptors; further, resorption liberates the ciliary basal body to differentiate into the centrosome, which performs discrete functions in S-, G2-, and M-phase. Besides these roles, the cilium provides a positional cue that regulates polarity of cell division, and thus directs cells towards fates of differentiation versus proliferation. In this review, we summarize the specific mechanisms mediating the cilia-cell cycle dialog. We then emphasize the examples of polycystic kidney disease (PKD), nephronopthisis (NPHP), and VHL-linked renal cysts as cases in which defects of ciliary function influence disease pathology, and may also condition response to treatment.
Collapse
Affiliation(s)
- Junmin Pan
- Protein Science Laboratory of the Ministry of Education, School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| | | | | |
Collapse
|
41
|
|
42
|
Lienkamp S, Ganner A, Walz G. Inversin, Wnt signaling and primary cilia. Differentiation 2012; 83:S49-55. [DOI: 10.1016/j.diff.2011.11.012] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 11/22/2011] [Accepted: 11/24/2011] [Indexed: 12/28/2022]
|
43
|
Werner ME, Mitchell BJ. Understanding ciliated epithelia: the power of Xenopus. Genesis 2011; 50:176-85. [PMID: 22083727 DOI: 10.1002/dvg.20824] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Revised: 11/04/2011] [Accepted: 11/08/2011] [Indexed: 01/20/2023]
Abstract
Ciliated epithelia are important in a wide variety of biological contexts where they generate directed fluid flow. Here we address the fundamental advances in understanding ciliated epithelia that have been achieved using Xenopus as a model system. Xenopus embryos are covered with a ciliated epithelium that propels fluid unidirectionally across their surface. The external nature of this tissue, coupled with the molecular tools available in Xenopus and the ease of microscopic analysis on intact animals has thrust Xenopus to the forefront of ciliated epithelia biology. We discuss advances in understanding the molecular regulators of ciliated epithelia cell fate as well as basic aspects of ciliated epithelia cell biology including ciliogenesis and cell polarity.
Collapse
Affiliation(s)
- M E Werner
- Department of Cell and Molecular Biology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60302, USA
| | | |
Collapse
|
44
|
Zhang B, Tran U, Wessely O. Expression of Wnt signaling components during Xenopus pronephros development. PLoS One 2011; 6:e26533. [PMID: 22028899 PMCID: PMC3197532 DOI: 10.1371/journal.pone.0026533] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2011] [Accepted: 09/28/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The formation of the vertebrate kidney is tightly regulated and relies on multiple evolutionarily conserved inductive events. These are present in the complex metanephric kidney of higher vertebrates, but also in the more primitive pronephric kidney functional in the larval stages of amphibians and fish. Wnts have long been viewed as central in this process. Canonical β-Catenin-dependent Wnt signaling establishes kidney progenitors and non-canonical β-Catenin-independent Wnt signaling participate in the morphogenetic processes that form the highly sophisticated nephron structure. While some individual Wnt signaling components have been studied extensively in the kidney, the overall pathway has not yet been analyzed in depth. METHODOLOGY/PRINCIPAL FINDINGS Here we report a detailed expression analysis of all Wnt ligands, receptors and several downstream Wnt effectors during pronephros development in Xenopus laevis using in situ hybridization. Out of 19 Wnt ligands, only three, Wnt4, Wnt9a and Wnt11, are specifically expressed in the pronephros. Others such as Wnt8a are present, but in a broader domain comprising adjacent tissues in addition to the kidney. The same paradigm is observed for the Wnt receptors and its downstream signaling components. Fzd1, Fzd4, Fzd6, Fzd7, Fzd8 as well as Celsr1 and Prickle1 show distinct expression domains in the pronephric kidney, whereas the non-traditional Wnt receptors, Ror2 and Ryk, as well as the majority of the effector molecules are rather ubiquitous. In addition to this spatial regulation, the timing of expression is also tightly regulated. In particular, non-canonical Wnt signaling seems to be restricted to later stages of pronephros development. CONCLUSION/SIGNIFICANCE Together these data suggest a complex cross talk between canonical and non-canonical Wnt signaling is required to establish a functional pronephric kidney.
Collapse
Affiliation(s)
- Bo Zhang
- Lerner Research Institute/Cleveland Clinic, Department of Cell Biology, Cleveland, Ohio, United States of America
- Louisiana State University (LSU) Health Sciences Center, Department of Cell Biology & Anatomy, New Orleans, Louisiana, United States of America
| | - Uyen Tran
- Lerner Research Institute/Cleveland Clinic, Department of Cell Biology, Cleveland, Ohio, United States of America
| | - Oliver Wessely
- Lerner Research Institute/Cleveland Clinic, Department of Cell Biology, Cleveland, Ohio, United States of America
- * E-mail:
| |
Collapse
|
45
|
Abstract
Planar cell polarity (PCP) describes the coordinated polarization of tissue cells in a direction that is orthogonal to their apical/basal axis. In the last several years, studies in flies and vertebrates have defined evolutionarily conserved pathways that establish and maintain PCP in various cellular contexts. Defective responses to the polarizing signal(s) have deleterious effects on the development and repair of a wide variety of organs/tissues. In this review, we cover the known and hypothesized roles for PCP in the metanephric kidney. We highlight the similarities and differences in PCP establishment in this organ compared with flies, especially the role of Wnt signaling in this process. Finally, we present a model whereby the signal(s) that organizes PCP in the kidney epithelium, at least in part, comes from the adjacent stromal fibroblasts.
Collapse
Affiliation(s)
- Thomas J Carroll
- Department of Internal Medicine (Nephrology), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | | |
Collapse
|
46
|
Nelson PJ, von Toerne C, Gröne HJ. Wnt-signaling pathways in progressive renal fibrosis. Expert Opin Ther Targets 2011; 15:1073-83. [PMID: 21623684 DOI: 10.1517/14728222.2011.588210] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION The prevention and potential reversal of interstitial fibrosis is a central strategy for the treatment of progressive renal disease. This strategy requires a better understanding of the underlying pathophysiologic processes involved in progressive renal fibrosis. AREAS COVERED The developmental processes in which Wnt (combination of 'wingless' and 'INT')/frizzled signaling is involved is discussed in this review, including cell fate determination, cell polarity, tissue patterning and control of cell proliferation. These pathways are also active in the adult where they play key roles in the maintenance of tissue homeostasis, wound repair and chronic tissue damage. EXPERT OPINION Wnt biology helps to control cell polarity, moderates cell proliferation and underlies other processes linked to renal homeostasis. Reactivation and dysregulation of the Wnt pathways underlie chronic fibrosis and progressive renal failure. Wnt signaling is, however, context-dependent: the pathways are complex and undergo many levels of cross-talk with other regulatory systems and regulatory pathways. On one hand, this may help to explain the positive effects of Wnt-signaling blockades seen in some animal models of chronic renal damage and, on the other, this suggests that it may be difficult to predict how modifications of the Wnt pathway may influence a process.
Collapse
Affiliation(s)
- Peter J Nelson
- Ludwig-Maximilians University, Medical Policlinic, Clinical Biochemistry Group, Munich, Germany.
| | | | | |
Collapse
|
47
|
Burcklé C, Gaudé HM, Vesque C, Silbermann F, Salomon R, Jeanpierre C, Antignac C, Saunier S, Schneider-Maunoury S. Control of the Wnt pathways by nephrocystin-4 is required for morphogenesis of the zebrafish pronephros. Hum Mol Genet 2011; 20:2611-27. [PMID: 21498478 DOI: 10.1093/hmg/ddr164] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nephronophthisis is a hereditary nephropathy characterized by interstitial fibrosis and cyst formation. It is caused by mutations in NPHP genes encoding the ciliary proteins, nephrocystins. In this paper, we investigate the function of nephrocystin-4, the product of the nphp4 gene, in vivo by morpholino-mediated knockdown in zebrafish and in vitro in mammalian kidney cells. Depletion of nephrocystin-4 results in convergence and extension defects, impaired laterality, retinal anomalies and pronephric cysts associated with alterations in early cloacal morphogenesis. These defects are accompanied by abnormal ciliogenesis in the cloaca and in the laterality organ. We show that nephrocystin-4 is required for the elongation of the caudal pronephric primordium and for the regulation of cell rearrangements during cloaca morphogenesis. Moreover, depletion of either inversin, the product of the nphp2 gene, or of the Wnt-planar cell polarity (PCP) pathway component prickle2 increases the proportion of cyst formation in nphp4-depleted embryos. Nephrocystin-4 represses the Wnt-β-catenin pathway in the zebrafish cloaca and in mammalian kidney cells in culture. In these cells, nephrocystin-4 interacts with inversin and dishevelled, and regulates dishevelled stability and subcellular localization. Our data point to a function of nephrocystin-4 in a tight regulation of the Wnt-β-catenin and Wnt-PCP pathways, in particular during morphogenesis of the zebrafish pronephros. Moreover, they highlight common signalling functions for inversin and nephrocystin-4, suggesting that these two nephrocystins are involved in common physiopathological mechanisms.
Collapse
Affiliation(s)
- Céline Burcklé
- INSERM U983, Tour Lavoisier, Hôpital Necker-Enfants Malades, 149 rue de Sèvres, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|