Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Sci Rep ; 13(1): 17647, 2023 10 17.
Article in English | MEDLINE | ID: mdl-37848494

ABSTRACT

CLIC5 belongs to a family of ion channels with six members reported so far. In vertebrates, the CLIC5 gene encodes two different isoforms, CLIC5A and CLIC5B. In addition to its ion channel activity, there is evidence for further functions of CLIC5A, such as the remodeling of the actin cytoskeleton during the formation of a functional glomerulus in the vertebrate kidney. However, its specific role is still incompletely understood and a specific functional role for CLIC5B has not been described yet. Here we report our findings on the differential expression and functions of Clic5a and Clic5b during zebrafish kidney development. Whole-mount in situ hybridization studies revealed specific expression of clic5a in the eye and pronephric glomerulus, and clic5b is expressed in the gut, liver and the pronephric tubules. Clic5 immunostainings revealed that Clic5b is localized in the cilia. Whereas knockdown of Clic5a resulted in leakiness of the glomerular filtration barrier, Clic5b deficient embryos displayed defective ciliogenesis, leading to ciliopathy-associated phenotypes such as ventral body curvature, otolith deposition defects, altered left-right asymmetry and formation of hydrocephalus and pronephric cysts. In addition, Clic5 deficiency resulted in dysregulation of cilia-dependent Wnt signalling pathway components. Mechanistically, we identified a Clic5-dependent activation of the membrane-cytoskeletal linker proteins Ezrin/Radixin/Moesin (ERM) in the pronephric tubules of zebrafish. In conclusion, our in vivo data demonstrates a novel role for Clic5 in regulating essential ciliary functions and identified Clic5 as a positive regulator of ERM phosphorylation.


Subject(s)
Chloride Channels , Chlorides , Cilia , Kidney Glomerulus , Microfilament Proteins , Zebrafish , Animals , Actin Cytoskeleton/metabolism , Chloride Channels/genetics , Chloride Channels/metabolism , Chlorides/metabolism , Cilia/genetics , Cilia/metabolism , Kidney Glomerulus/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Zebrafish/genetics , Zebrafish/metabolism
2.
Biochem Biophys Res Commun ; 584: 19-25, 2021 12 20.
Article in English | MEDLINE | ID: mdl-34753064

ABSTRACT

The primary cilium is a sensory organelle at the cell surface with integral functions in cell signaling. It contains a microtubular axoneme that is rooted in the basal body (BB) and serves as a scaffold for the movement of intraflagellar transport (IFT) particles by Kinesin-2 along the cilium. Ift88, a member of the anterograde moving IFT-B1 complex, as well as the Kinesin-2 subunit Kif3a are required for cilia formation. To facilitate signaling, the cilium restricts the access of molecules to its membrane ("ciliary gate"). This is thought to be mediated by cytoskeletal barriers ("subciliary domains") originating from the BB subdistal/distal appendages, the periciliary membrane compartment (PCMC) as well as the transition fibers and zone (TF/TZ). The PCMC is a poorly characterized membrane domain surrounding the ciliary base with exclusion of certain apical membrane proteins. Here we describe that Ift88, but not Kinesin-2, is required for the establishment of the PCMC in MDCK cells. Likewise, in C. elegans mutants of the Ift88 ortholog osm-5 fail to establish the PCMC, while Kinesin-2 deficient osm-3 mutants form PCMCs normally. Furthermore, disruption of IFT-B1 into two subcomplexes, while disrupting ciliogenesis, does not interfere with PCMC formation. Our findings suggest that cilia are not a prerequisite for the formation of the PCMC, and that separate machineries with partially overlapping functions are required for the establishment of each.


Subject(s)
Cell Membrane/metabolism , Cilia/metabolism , Epithelial Cells/metabolism , Kinesins/metabolism , Membrane Transport Proteins/metabolism , Animals , Basal Bodies/metabolism , Caenorhabditis elegans/cytology , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/metabolism , Cytoskeleton/metabolism , Dogs , Madin Darby Canine Kidney Cells , Microscopy, Fluorescence , Nerve Tissue Proteins/metabolism , Signal Transduction
3.
Nat Commun ; 9(1): 3660, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30202007

ABSTRACT

Kidney injury is a common complication of severe disease. Here, we report that injuries of the zebrafish embryonal kidney are rapidly repaired by a migratory response in 2-, but not in 1-day-old embryos. Gene expression profiles between these two developmental stages identify cxcl12a and myca as candidates involved in the repair process. Zebrafish embryos with cxcl12a, cxcr4b, or myca deficiency display repair abnormalities, confirming their role in response to injury. In mice with a kidney-specific knockout, Cxcl12 and Myc gene deletions suppress mitochondrial metabolism and glycolysis, and delay the recovery after ischemia/reperfusion injury. Probing these observations in zebrafish reveal that inhibition of glycolysis slows fast migrating cells and delays the repair after injury, but does not affect the slow cell movements during kidney development. Our findings demonstrate that Cxcl12 and Myc facilitate glycolysis to promote fast migratory responses during development and repair, and potentially also during tumor invasion and metastasis.


Subject(s)
Chemokine CXCL12/metabolism , Gene Expression Regulation, Developmental , Kidney Diseases/metabolism , Proto-Oncogene Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Animals, Genetically Modified , Cell Movement , Energy Metabolism , Gene Deletion , Gene Expression Profiling , Glycolysis , Homeostasis , Kidney/injuries , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction , Tretinoin/chemistry
4.
PLoS Biol ; 16(8): e2005651, 2018 08.
Article in English | MEDLINE | ID: mdl-30080851

ABSTRACT

Cilia are organelles specialized in movement and signal transduction. The ciliary transient receptor potential ion channel polycystin-2 (TRPP2) controls elementary cilia-mediated physiological functions ranging from male fertility and kidney development to left-right patterning. However, the molecular components translating TRPP2 channel-mediated Ca2+ signals into respective physiological functions are unknown. Here, we show that the Ca2+-regulated mitochondrial ATP-Mg/Pi solute carrier 25 A 25 (SLC25A25) acts downstream of TRPP2 in an evolutionarily conserved metabolic signaling pathway. We identify SLC25A25 as an essential component in this cilia-dependent pathway using a genome-wide forward genetic screen in Drosophila melanogaster, followed by a targeted analysis of SLC25A25 function in zebrafish left-right patterning. Our data suggest that TRPP2 ion channels regulate mitochondrial SLC25A25 transporters via Ca2+ establishing an evolutionarily conserved molecular link between ciliary signaling and mitochondrial metabolism.


Subject(s)
Amino Acid Transport Systems, Acidic/metabolism , Calcium-Binding Proteins/metabolism , Cilia/metabolism , TRPP Cation Channels/metabolism , Animals , Antiporters/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Drosophila melanogaster/metabolism , Heterozygote , Humans , Ion Channels/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Proteins/metabolism , Signal Transduction , Zebrafish
5.
J Biol Chem ; 293(39): 15243-15255, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30111592

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.


Subject(s)
Ciliopathies/genetics , Kidney Diseases, Cystic/congenital , NM23 Nucleoside Diphosphate Kinases/genetics , Renal Insufficiency/genetics , Animals , Cell Cycle Checkpoints/genetics , Cilia/genetics , Cilia/pathology , Ciliopathies/physiopathology , DNA Damage/genetics , DNA Repair/genetics , Disease Models, Animal , Humans , Kidney/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Microtubule Proteins/genetics , NIMA-Related Kinases/genetics , Nuclear Proteins/genetics , Renal Insufficiency/pathology , Xenopus/genetics , Zebrafish/genetics
6.
Proc Natl Acad Sci U S A ; 114(23): E4621-E4630, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28536193

ABSTRACT

Podocytes form the outer part of the glomerular filter, where they have to withstand enormous transcapillary filtration forces driving glomerular filtration. Detachment of podocytes from the glomerular basement membrane precedes most glomerular diseases. However, little is known about the regulation of podocyte adhesion in vivo. Thus, we systematically screened for podocyte-specific focal adhesome (FA) components, using genetic reporter models in combination with iTRAQ-based mass spectrometry. This approach led to the identification of FERM domain protein EPB41L5 as a highly enriched podocyte-specific FA component in vivo. Genetic deletion of Epb41l5 resulted in severe proteinuria, detachment of podocytes, and development of focal segmental glomerulosclerosis. Remarkably, by binding and recruiting the RhoGEF ARGHEF18 to the leading edge, EPB41L5 directly controls actomyosin contractility and subsequent maturation of focal adhesions, cell spreading, and migration. Furthermore, EPB41L5 controls matrix-dependent outside-in signaling by regulating the focal adhesome composition. Thus, by linking extracellular matrix sensing and signaling, focal adhesion maturation, and actomyosin activation EPB41L5 ensures the mechanical stability required for podocytes at the kidney filtration barrier. Finally, a diminution of EPB41L5-dependent signaling programs appears to be a common theme of podocyte disease, and therefore offers unexpected interventional therapeutic strategies to prevent podocyte loss and kidney disease progression.


Subject(s)
Actomyosin/metabolism , Cytoskeletal Proteins/metabolism , Focal Adhesions/metabolism , Membrane Proteins/metabolism , Podocytes/metabolism , Animals , Cytoskeletal Proteins/deficiency , Cytoskeletal Proteins/genetics , Female , Focal Adhesions/pathology , Gene Knockout Techniques , Glomerulosclerosis, Focal Segmental/etiology , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Nephrotic Syndrome/etiology , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Podocytes/pathology , Pregnancy , Proteomics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction
7.
Transplant Direct ; 2(4): e70, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27500261

ABSTRACT

UNLABELLED: Calciphylaxis is a rare and often fatal condition mostly associated with end-stage renal disease. The pathophysiology remains elusive and treatment options are scarce. We present a rare case of severe calciphylaxis after kidney transplantation in a patient with persistent hyperparathyroidism. CASE DESCRIPTION: A 78-year-old man with a history of end-stage renal disease developed edema and ulcerations on both lower limbs 14 months after kidney transplantation while receiving an mammalian target of rapamycin inhibitor to manage polyoma virus-associated nephropathy. Skin biopsies taken from the ulcerations confirmed calciphylaxis. A multimodal treatment regimen combining medical (calcium-free phosphate binders, cinacalcet, paricalcitol, sodium thiosulfate, antibiotic treatment) and surgical treatments (debridement and autologous skin transplantation) ultimately resulted in successful wound healing. DISCUSSION: We describe a case of severe calciphylaxis in a nonuremic patient after kidney transplantation. Rapid diagnosis by skin biopsy and an aggressive multimodal therapy regimen followed by long-term oral sodium thiosulfate treatment were crucial factors for a favorable outcome.

8.
Nephrol Dial Transplant ; 31(2): 262-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26420894

ABSTRACT

BACKGROUND: Chronic kidney disease (CKD) in children is characterized by rapid progression and a high incidence of end-stage renal disease and therefore constitutes an important health problem. While unbiased genetic screens have identified common risk variants influencing renal function and CKD in adults, the presence and identity of such variants in pediatric CKD are unknown. METHODS: The international Pediatric Investigation for Genetic Factors Linked with Renal Progression (PediGFR) Consortium comprises three pediatric CKD cohorts: Chronic Kidney Disease in Children (CKiD), Effect of Strict Blood Pressure Control and ACE Inhibition on the Progression of CRF in Pediatric Patients (ESCAPE) and Cardiovascular Comorbidity in Children with CKD (4C). Clean genotype data from > 10 million genotyped or imputed single-nucleotide polymorphisms (SNPs) were available for 1136 patients with measurements of serum creatinine at study enrollment. Genome-wide association studies were conducted to relate the SNPs to creatinine-based estimated glomerular filtration rate (eGFR crea) and proteinuria (urinary albumin- or protein-to-creatinine ratio ≥ 300 and ≥ 500 mg/g, respectively). In addition, European-ancestry PediGFR patients (cases) were compared with 1347 European-ancestry children without kidney disease (controls) to identify genetic variants associated with the presence of CKD. RESULTS: SNPs with suggestive association P-values < 1 × 10(-5) were identified in 10 regions for eGFR crea, four regions for proteinuria and six regions for CKD including some plausible biological candidates. No SNP was associated at genome-wide significance (P < 5 × 10(-8)). Investigation of the candidate genes for proteinuria in adults from the general population provided support for a region on chromosome 15 near RSL24D1/UNC13C/RAB27A. Conversely, targeted investigation of genes harboring GFR-associated variants in adults from the general population did not reveal significantly associated SNPs in children with CKD. CONCLUSIONS: Our findings suggest that larger collaborative efforts will be needed to draw reliable conclusions about the presence and identity of common variants associated with eGFR, proteinuria and CKD in pediatric populations.


Subject(s)
Genome-Wide Association Study/methods , Glomerular Filtration Rate/physiology , Polymorphism, Single Nucleotide , Renal Insufficiency, Chronic/genetics , Adolescent , Child , Child, Preschool , Disease Progression , Europe/epidemiology , Female , Genetic Loci , Genotype , Humans , Infant , Male , Morbidity/trends , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/physiopathology , Risk Factors
9.
Nephrol Dial Transplant ; 31(4): 663-71, 2016 04.
Article in English | MEDLINE | ID: mdl-26610596

ABSTRACT

BACKGROUND: ABO-incompatible kidney transplantation (ABOi KTx) expands the living donor transplantation options. However, long-term outcome data, especially in comparison with ABO-compatible kidney transplantation (ABOc KTx), remain limited. Since the first ABOi KTx in Germany on 1 April 2004 at our centre, we have followed 100 ABOi KTx over up to 10 years. METHODS: One hundred ABOi KTx and 248 ABOc KTx from 1 April 2004 until 28 October 2014 were analysed in this observational, single-centre study. Three ABOi KTx and 141 ABOc KTx were excluded because of cyclosporine A-based immunosuppression, and 1 ABOc KTx was lost to follow-up. RESULTS: Median estimated 10-year patient and graft survival in ABOi KTx was 99 and 94%, respectively, and surpassed ABOc-KTx patient and graft survival of 80 and 88%, respectively. The incidence rate of antibody-mediated rejections was 10 and 8%, and that of T-cell-mediated rejections was 17 and 20% in ABOi KTx and ABOc KTx, respectively. Infectious and malignant complications in ABOi KTx were not more common than in ABOc KTx. However, postoperative lymphoceles occurred more frequently in ABOi KTx. Subgroup analysis of ABOi-KTx patients revealed that patients with high-titre isohaemagglutinins before transplantation had equal long-term results compared with low-titre isohaemagglutinin patients. CONCLUSION: Taken together, long-term outcome of ABOi KTx is not inferior to ABOc KTx. Incidences of rejection episodes, infectious complications and malignancies are not increased, despite the more vigorous immunosuppression in ABOi KTx. Our data provide further evidence that ABOi KTx with living donation is a safe, successful and reasonable option to reduce the organ shortage.


Subject(s)
ABO Blood-Group System/immunology , Blood Group Incompatibility/immunology , Graft Rejection/epidemiology , Infections/epidemiology , Kidney Transplantation , Adolescent , Adult , Aged , Female , Germany/epidemiology , Graft Survival , Humans , Immunosuppression Therapy , Male , Middle Aged , Postoperative Complications , Prospective Studies , Time Factors , Young Adult
11.
Transpl Int ; 28(4): 387-97, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25387763

ABSTRACT

ABO-incompatible kidney transplantation is nowadays a well-established procedure to expand living donor transplantation to blood group incompatible donor/recipient constellations. In the last two decades, transplantation protocols evolved to more specific isohaemagglutinin elimination techniques and established competent antirejection protection protocols without the need of splenectomy. ABOi kidney transplantation associated accommodation despite isohaemagglutinin reappearance, C4d positivity of peritubular capillaries as well as the increased incidence of bleeding complications is currently under intense investigation. However, most recent data show excellent graft survival rates equivalent to ABO-compatible kidney transplantation outcome.


Subject(s)
ABO Blood-Group System , Kidney Transplantation , Transplantation Immunology , Clinical Protocols , Complement C4/immunology , Graft Rejection/prevention & control , Hemagglutinins/blood , Hemagglutinins/isolation & purification , Humans , Immunoglobulins, Intravenous , Immunologic Factors/therapeutic use , Rituximab/therapeutic use
12.
Development ; 142(1): 174-84, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25516973

ABSTRACT

Cilia are microtubule-based organelles that are present on most cells and are required for normal tissue development and function. Defective cilia cause complex syndromes with multiple organ manifestations termed ciliopathies. A crucial step during ciliogenesis in multiciliated cells (MCCs) is the association of future basal bodies with the apical plasma membrane, followed by their correct spacing and planar orientation. Here, we report a novel role for ELMO-DOCK1, which is a bipartite guanine nucleotide exchange factor complex for the small GTPase Rac1, and for the membrane-cytoskeletal linker Ezrin, in regulating centriole/basal body migration, docking and spacing. Downregulation of each component results in ciliopathy-related phenotypes in zebrafish and disrupted ciliogenesis in Xenopus epidermal MCCs. Subcellular analysis revealed a striking impairment of basal body docking and spacing, which is likely to account for the observed phenotypes. These results are substantiated by showing a genetic interaction between elmo1 and ezrin b. Finally, we provide biochemical evidence that the ELMO-DOCK1-Rac1 complex influences Ezrin phosphorylation and thereby probably serves as an important molecular switch. Collectively, we demonstrate that the ELMO-Ezrin complex orchestrates ciliary basal body migration, docking and positioning in vivo.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Basal Bodies/metabolism , Cilia/metabolism , Cytoskeletal Proteins/metabolism , Xenopus Proteins/metabolism , Zebrafish Proteins/metabolism , rac1 GTP-Binding Protein/metabolism , Animals , Axoneme/metabolism , Axoneme/ultrastructure , Cell Membrane/metabolism , Cilia/ultrastructure , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/ultrastructure , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Models, Biological , Phosphorylation , Protein Binding , Xenopus laevis , Zebrafish/embryology , rac GTP-Binding Proteins
13.
Case Rep Med ; 2014: 613641, 2014.
Article in English | MEDLINE | ID: mdl-25317177

ABSTRACT

We present a case report of a 59-year-old man, who received a blood group identical living unrelated kidney graft. This was his second kidney transplantation. Pretransplant T-cell crossmatch resulted negative. B-cell crossmatch, which is not considered a strict contraindication for transplantation, resulted positive. During surgery no abnormalities occurred. Four hours after the transplantation diuresis suddenly decreased. In an immediately performed relaparotomy the transplanted kidney showed signs of hyperacute rejection and had to be removed. Pathological examination was consistent with hyperacute rejection. Depositions of IgM or IgG antibodies were not present in pathologic evaluation of the rejected kidney, suggesting that no irregular endothelial specific antibodies had been involved in the rejection. We recommend examining more closely recipients of second allografts, considering not only a positive T-cell crossmatch but also a positive B-cell crossmatch as exclusion criteria for transplantation.

14.
J Biol Chem ; 289(12): 8390-401, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24500717

ABSTRACT

Although the two ciliopathies Bardet-Biedl syndrome and nephronophthisis share multiple clinical manifestations, the molecular basis for this overlap remains largely unknown. Both BBS11 and NPHP7 are unusual members of their respective gene families. Although BBS11/TRIM32 represents a RING finger E3 ubiquitin ligase also involved in hereditary forms of muscular dystrophy, NPHP7/Glis2 is a Gli-like transcriptional repressor that localizes to the nucleus, deviating from the ciliary localization of most other ciliopathy-associated gene products. We found that BBS11/TRIM32 and NPHP7/Glis2 can physically interact with each other, suggesting that both proteins form a functionally relevant protein complex in vivo. This hypothesis was further supported by the genetic interaction and synergist cyst formation in the zebrafish pronephros model. However, contrary to our expectation, the E3 ubiquitin ligase BBS11/TRIM32 was not responsible for the short half-life of NPHP7/Glis2 but instead promoted the accumulation of mixed Lys(48)/Lys(63)-polyubiquitylated NPHP7/Glis2 species. This modification not only prolonged the half-life of NPHP7/Glis2, but also altered the subnuclear localization and the transcriptional activity of NPHP7/Glis2. Thus, physical and functional interactions between NPHP and Bardet-Biedl syndrome gene products, demonstrated for Glis2 and TRIM32, may help to explain the phenotypic similarities between these two syndromes.


Subject(s)
Bardet-Biedl Syndrome/metabolism , Kidney Diseases, Cystic/metabolism , Kruppel-Like Transcription Factors/metabolism , Transcription Factors/metabolism , Animals , Bardet-Biedl Syndrome/genetics , HEK293 Cells , Humans , Kidney Diseases, Cystic/genetics , Kruppel-Like Transcription Factors/analysis , Kruppel-Like Transcription Factors/genetics , Protein Interaction Maps , Protein Transport , Transcription Factors/genetics , Transcriptional Activation , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Zebrafish
15.
PLoS One ; 8(9): e72549, 2013.
Article in English | MEDLINE | ID: mdl-24069149

ABSTRACT

Bardet-Biedl syndrome (BBS) and nephronophthisis (NPH) are hereditary autosomal recessive disorders, encoded by two families of diverse genes. BBS and NPH display several overlapping phenotypes including cystic kidney disease, retinitis pigmentosa, liver fibrosis, situs inversus and cerebellar defects. Since most of the BBS and NPH proteins localize to cilia and/or their appendages, BBS and NPH are considered ciliopathies. In this study, we characterized the function of the transcription factor Nphp7 in zebrafish, and addressed the molecular connection between BBS and NPH. The knockdown of zebrafish bbs1 and nphp7.2 caused similar phenotypic changes including convergent extension defects, curvature of the body axis, hydrocephalus, abnormal heart looping and cystic pronephros, all consistent with an altered ciliary function. Immunoprecipitation assays revealed a physical interaction between BBS1 and NPHP7, and the simultaneous knockdown of zbbs1 and znphp7.2 enhanced the cystic pronephros phenotype synergistically, suggesting a genetic interaction between zbbs1 and znphp7.2 in vivo. Deletion of zBbs1 or zNphp7.2 did not compromise cilia formation, but disrupted cilia motility. Although NPHP7 has been shown to act as transcriptional repressor, our studies suggest a crosstalk between BBS1 and NPHP7 in regulating normal function of the cilium.


Subject(s)
Cilia/metabolism , Cilia/physiology , Nuclear Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Nuclear Proteins/genetics , Protein Binding , Zebrafish , Zebrafish Proteins/genetics
16.
Nat Genet ; 45(8): 951-6, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23793029

ABSTRACT

Nephronophthisis is an autosomal recessive cystic kidney disease that leads to renal failure in childhood or adolescence. Most NPHP gene products form molecular networks. Here we identify ANKS6 as a new NPHP family member that connects NEK8 (NPHP9) to INVS (NPHP2) and NPHP3. We show that ANKS6 localizes to the proximal cilium and confirm its role in renal development through knockdown experiments in zebrafish and Xenopus laevis. We also identify six families with ANKS6 mutations affected by nephronophthisis, including severe cardiovascular abnormalities, liver fibrosis and situs inversus. The oxygen sensor HIF1AN hydroxylates ANKS6 and INVS and alters the composition of the ANKS6-INVS-NPHP3 module. Knockdown of Hif1an in Xenopus results in a phenotype that resembles loss of other NPHP proteins. Network analyses uncovered additional putative NPHP proteins and placed ANKS6 at the center of this NPHP module, explaining the overlapping disease manifestation caused by mutation in ANKS6, NEK8, INVS or NPHP3.


Subject(s)
Kidney Diseases, Cystic/genetics , Kinesins/genetics , Nuclear Proteins/genetics , Protein Kinases/genetics , Transcription Factors/genetics , Animals , Cilia/metabolism , Consanguinity , Exons , Gene Knockdown Techniques , Humans , Introns , Kidney Diseases, Cystic/metabolism , Kinesins/metabolism , Mice , Mutation , NIMA-Related Kinases , Nuclear Proteins/metabolism , Phenotype , Polycystic Kidney Diseases/genetics , Protein Binding , Protein Interaction Maps , Protein Kinases/metabolism , Protein Transport , Transcription Factors/metabolism , Xenopus/embryology , Xenopus/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
17.
Nat Genet ; 44(6): 714-9, 2012 May 13.
Article in English | MEDLINE | ID: mdl-22581229

ABSTRACT

Cilia are essential for fertilization, respiratory clearance, cerebrospinal fluid circulation and establishing laterality. Cilia motility defects cause primary ciliary dyskinesia (PCD, MIM244400), a disorder affecting 1:15,000-30,000 births. Cilia motility requires the assembly of multisubunit dynein arms that drive ciliary bending. Despite progress in understanding the genetic basis of PCD, mutations remain to be identified for several PCD-linked loci. Here we show that the zebrafish cilia paralysis mutant schmalhans (smh(tn222)) encodes the coiled-coil domain containing 103 protein (Ccdc103), a foxj1a-regulated gene product. Screening 146 unrelated PCD families identified individuals in six families with reduced outer dynein arms who carried mutations in CCDC103. Dynein arm assembly in smh mutant zebrafish was rescued by wild-type but not mutant human CCDC103. Chlamydomonas Ccdc103/Pr46b functions as a tightly bound, axoneme-associated protein. These results identify Ccdc103 as a dynein arm attachment factor that causes primary ciliary dyskinesia when mutated.


Subject(s)
Dyneins/metabolism , Kartagener Syndrome/genetics , Animals , Cilia/metabolism , Female , Humans , Male , Mutation , Pedigree , Zebrafish
18.
Hum Mol Genet ; 20(16): 3119-28, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21596840

ABSTRACT

NPHP4 mutations cause nephronophthisis, an autosomal recessive cystic kidney disease associated with renal fibrosis and kidney failure. The NPHP4 gene product nephrocystin-4 interacts with other nephrocystins, cytoskeletal and ciliary proteins; however, the molecular and cellular functions of nephrocystin-4 have remained elusive. Here we demonstrate that nephrocystin-4 is required for normal cloaca formation during zebrafish embryogenesis. Time-lapse imaging of the developing zebrafish pronephros revealed that tubular epithelial cells at the distal pronephros actively migrate between the yolk sac extension and the blood island towards the ventral fin fold to join the proctodeum and to form the cloaca. Nphp4-deficient pronephric duct cells failed to connect with their ectodermal counterparts, and instead formed a vesicle at the obstructed end of the pronephric duct. Nephrocystin-4 interacts with nephrocystin-1 and Par6. Depletion of zebrafish NPHP1 (nphp1) increased the incidence of cyst formation and randomization of the normal body axis, but did not augment cloaca malformation in nphp4-deficient zebrafish embryos. However, simultaneous depletion of zebrafish Par6 (pard6) aggravated cloaca formation defects in nphp4-depleted embryos, suggesting that nphp4 orchestrates directed cell migration and cloaca formation through interaction with the Par protein complex.


Subject(s)
Cloaca/embryology , Nephrons/embryology , Zebrafish Proteins/metabolism , Zebrafish/embryology , Amino Acid Sequence , Animals , Cell Movement , Cilia/metabolism , Cloaca/metabolism , Cloaca/pathology , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/pathology , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Molecular Sequence Data , Nephrons/metabolism , Nephrons/pathology , Phenotype , Zebrafish/genetics , Zebrafish Proteins/chemistry , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
19.
Hum Mol Genet ; 19(12): 2347-59, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20233749

ABSTRACT

Mutations of the immunoglobulin superfamily proteins nephrin and Neph1 lead to congenital nephrotic syndrome in humans or mice. Neph proteins are three closely related molecules that are evolutionarily conserved and mediate cell recognition. Their importance for morphogenetic processes including the formation of the kidney filtration barrier in vertebrates and synaptogenesis in Caenorhabditis elegans has recently been uncovered. However, the individual morphogenetic function of mammalian Neph1-3 isoforms remained elusive. We demonstrate now that the Neph/nephrin family proteins can form cell-cell adhesion modules across species. Expression of all three mammalian Neph isoforms partially rescued mutant C. elegans lacking their Neph homolog syg-1 and restored synapse formation, suggesting a functional redundancy between the three isoforms. Strikingly, the rescue of defective synaptic connectivity was prevented by deletion of the highly conserved cytoplasmic PSD95/Dlg/ZO-1-binding motif of SYG-1/Neph proteins, indicating the critical role of this intracellular signaling motif for SYG-1/Neph-dependent morphogenetic events. To determine the significance of Neph isoform redundancy for vertebrate kidney development, we analyzed the expression pattern and the functional role of Neph proteins in zebrafish. In situ hybridizations identified zNeph1 and zNeph2 as glomerular proteins. Morpholino knockdown of either zNeph1 or zNeph2 resulted in loss of slit diaphragms and leakiness of the glomerular filtration barrier. This is the first report utilizing C. elegans to study mammalian Neph/nephrin protein function and to demonstrate a functional overlap of Neph1-3 proteins. Furthermore, we identify Neph2 as a novel critical regulator of glomerular function, indicating that both Neph1 and Neph2 are required for glomerular maintenance and development.


Subject(s)
Immunoglobulins/physiology , Kidney/growth & development , Membrane Proteins/physiology , Neurons/physiology , Amino Acid Sequence , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/growth & development , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/classification , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans Proteins/physiology , Cell Adhesion , HeLa Cells , Humans , Immunoglobulins/classification , Immunoglobulins/genetics , Membrane Proteins/classification , Membrane Proteins/genetics , Mice , Models, Animal , Morphogenesis/genetics , PDZ Domains , Phylogeny
20.
Hum Mol Genet ; 19(1): 16-24, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19801576

ABSTRACT

Autosomal dominant polycystic liver disease (PCLD) is caused by mutations of either PRKCSH or Sec63, two proteins associated with the endoplasmic reticulum (ER). Both proteins are involved in carbohydrate processing, folding and translocation of newly synthesized glycoproteins. It is postulated that defective quality control of proteins initiates endoplasmic reticulum-associated degradation (ERAD), which disrupts hepatic homeostasis in patients with PRKCSH or Sec63 mutations. However, the precise molecular mechanisms are not known. Here, we show that over-expression or depletion of PRKCSH in zebrafish embryos leads to pronephric cysts, abnormal body curvature and situs inversus. Identical phenotypic changes are induced by depletion or over-expression of TRPP2. Increased PRKCSH levels ameliorate developmental abnormalities caused by over-expressed TRPP2, whereas excess TRPP2 can compensate the loss PRKCSH, indicating that the proteins share a common signaling pathway. PRKCSH binds the C-terminal domain of TRPP2, and both proteins co-localize within the ER. Furthermore, PRKCSH interacts with Herp, and inhibits Herp-mediated ubiquitination of TRPP2. Our findings suggest that PRKCSH functions as a chaperone-like molecule, which prevents ERAD of TRPP2. Dysequilibrium between TRPP2 and PRKCSH may lead to cyst formation in PCLD patients with PRKCSH mutations, and thereby account for the overlapping manifestations observed in PCLD and autosomal dominant polycystic kidney disease.


Subject(s)
Carrier Proteins/metabolism , Molecular Chaperones/metabolism , Mutation/genetics , Polycystic Kidney, Autosomal Dominant/metabolism , Protein Processing, Post-Translational , TRPP Cation Channels/metabolism , Ubiquitins/metabolism , Zebrafish Proteins/metabolism , Animals , Carrier Proteins/genetics , Dogs , Embryo, Nonmammalian/abnormalities , Embryo, Nonmammalian/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques , Humans , Molecular Chaperones/genetics , Nephrons/drug effects , Nephrons/metabolism , Nephrons/pathology , Oligonucleotides, Antisense/pharmacology , Protein Binding/drug effects , Protein Processing, Post-Translational/drug effects , TRPP Cation Channels/genetics , Ubiquitination/drug effects , Ubiquitins/genetics , Zebrafish/embryology , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...