Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
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.
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
3.
J Cell Biol ; 211(5): 963-73, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26644512

ABSTRACT

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.


Subject(s)
Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Cilia/metabolism , Microfilament Proteins/metabolism , Proteins/metabolism , Xenopus Proteins/metabolism , Actin Cytoskeleton/metabolism , Animals , Basal Bodies/metabolism , Drosophila melanogaster , Epidermis/metabolism , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Immunoprecipitation , Membrane Proteins/metabolism , Molecular Sequence Data , Oligonucleotides/chemistry , Protein Binding , Protein Structure, Tertiary , Xenopus laevis/metabolism , rho GTP-Binding Proteins
5.
Kidney Int ; 87(6): 1191-200, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25671767

ABSTRACT

Nephronophthisis (NPH) is a heterogenetic autosomal recessive disorder associated with kidney cysts and multiple extrarenal manifestations. The disease-associated gene products (NPHPs) typically contain domains involved in protein-protein interactions, and appear to exert their tissue-specific functions in large protein complexes. Most NPHPs localize to the cilium and/or basal body; however, their precise molecular functions remain largely unknown. We have recently identified the SAM-domain containing protein Anks3 as a potential ANKS6/NPHP16-interacting protein, and report now that Anks3 interacts with several NPHPs as well as with Bicc1 and the oxygen-sensitive asparaginyl hydroxylase HIF1AN. Knockdown of anks3 in zebrafish embryos was associated with NPH-typical manifestations, including ciliary abnormalities, cyst formation, and laterality defects. In multi-ciliated epidermal cells, GFP-tagged Anks3 localizes to the cilium, but forms large aggregates in the absence of NPHP1, indicating that the negatively charged NPHP1 curtails the polymerization of Anks3. Collectively, these findings suggest that Anks3 is a cilia-associated molecule that partners with the ANKS6- and via NPHP1 to the NPHP1-4-8 module. Thus, developmental defects associated with Anks3 depletion in zebrafish suggest that ANKS3 mutations may cause NPH or NPH-like disease in humans.


Subject(s)
Carrier Proteins/genetics , Carrier Proteins/metabolism , Kidney/embryology , Xenopus Proteins/metabolism , Zebrafish Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cilia/metabolism , Cytoskeletal Proteins , Gene Knockdown Techniques , HEK293 Cells , Humans , Kidney/abnormalities , Kidney Diseases, Cystic/metabolism , Membrane Proteins/metabolism , Mixed Function Oxygenases/metabolism , NIMA-Related Kinases , Nuclear Proteins/metabolism , Polymerization , Protein Kinases/metabolism , Proteins/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Situs Inversus/genetics , Xenopus , Xenopus Proteins/genetics , Zebrafish , Zebrafish Proteins/metabolism
6.
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
7.
J Biol Chem ; 289(38): 26344-26356, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25100726

ABSTRACT

Tight regulation of Wnt/ß-catenin signaling is critical for vertebrate development and tissue maintenance, and deregulation can lead to a host of disease phenotypes, including developmental disorders and cancer. Proteins associated with primary cilia and centrosomes have been demonstrated to negatively regulate canonical Wnt signaling in interphase cells. The plant homeodomain zinc finger protein Jade-1 can act as an E3 ubiquitin ligase-targeting ß-catenin for proteasomal degradation and concentrates at the centrosome and ciliary basal body in addition to the nucleus in interphase cells. We demonstrate that the destruction complex component casein kinase 1α (CK1α) phosphorylates Jade-1 at a conserved SLS motif and reduces the ability of Jade-1 to inhibit ß-catenin signaling. Consistently, Jade-1 lacking the SLS motif is more effective than wild-type Jade-1 in reducing ß-catenin-induced secondary axis formation in Xenopus laevis embryos in vivo. Interestingly, CK1α also phosphorylates ß-catenin and the destruction complex component adenomatous polyposis coli at a similar SLS motif to the effect that ß-catenin is targeted for degradation. The opposing effect of Jade-1 phosphorylation by CK1α suggests a novel example of the dual functions of CK1α activity to either oppose or promote canonical Wnt signaling in a context-dependent manner.


Subject(s)
Casein Kinase Ialpha/physiology , Homeodomain Proteins/metabolism , Protein Processing, Post-Translational , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Conserved Sequence , Enzyme Repression , Gene Expression , HEK293 Cells , Humans , Molecular Sequence Data , Phosphorylation , Wnt Signaling Pathway , Xenopus laevis , beta Catenin/metabolism
8.
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
9.
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
10.
Development ; 139(16): 2966-77, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22764054

ABSTRACT

Carotenoids and their metabolites are widespread and exert key biological functions in living organisms. In vertebrates, the carotenoid oxygenase BCMO1 converts carotenoids such as ß,ß-carotene to retinoids, which are required for embryonic pattern formation and cell differentiation. Vertebrate genomes encode a structurally related protein named BCDO2 but its physiological function remains undefined. Here, we show that BCDO2 is expressed as an oxidative stress-regulated protein during zebrafish development. Targeted knockdown of this mitochondrial enzyme resulted in anemia at larval stages. Marker gene analysis and staining for hemoglobin revealed that erythropoiesis was not impaired but that erythrocytes underwent apoptosis in BCDO2-deficient larvae. To define the mechanism of this defect, we have analyzed the role of BCDO2 in human cell lines. We found that carotenoids caused oxidative stress in mitochondria that eventually led to cytochrome c release, proteolytic activation of caspase 3 and PARP1, and execution of the apoptotic pathway. Moreover, BCDO2 prevented this induction of the apoptotic pathway by carotenoids. Thus, our study identifying BCDO2 as a crucial protective component against oxidative stress establishes this enzyme as mitochondrial carotenoid scavenger and a gatekeeper of the intrinsic apoptotic pathway.


Subject(s)
Apoptosis/physiology , Carotenoids/metabolism , Mitochondria/metabolism , Oxygenases/metabolism , Zebrafish Proteins/metabolism , Acetylcysteine/pharmacology , Anemia/enzymology , Anemia/genetics , Animals , Apoptosis/drug effects , Base Sequence , Carotenoids/pharmacology , Cell Line , Dioxygenases , Erythropoiesis/genetics , Erythropoiesis/physiology , Fatty Acid Desaturases/genetics , Fatty Acid Desaturases/metabolism , Fenretinide/pharmacology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Gene Knockdown Techniques , Humans , Models, Biological , Oxidative Stress , Oxygenases/antagonists & inhibitors , Oxygenases/genetics , RNA, Small Interfering/genetics , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics
11.
Int J Pharm ; 410(1-2): 75-82, 2011 May 30.
Article in English | MEDLINE | ID: mdl-21421034

ABSTRACT

DNAzymes are a group of RNA-cleaving DNA oligonucleotides that contain a catalytic domain and represent a novel class of antisense molecules. Although single-stranded DNAzymes may represent the most effective nucleic acid drug to date, the sensitivity to nuclease degradation is challenging. Therefore, it is important to develop a drug delivery system, which protects the molecule against degradation during dermal application. In the present study, the potential protective effect, regarding the dermal application of DNAzyme, of multiple (W/O/W) emulsions, W/O emulsions, submicron emulsion and microemulsions were investigated using a HPLC method. The HPLC method enables the quantitative analysis of DNAzyme as well as the detection of degradation products. The differences between the activity of DNase I and the activity of nucleases located in the porcine skin were compared. It was found that the degradation of an aqueous solution of DNAzyme is depending on the DNase I activity as well as on the incubation time. Furthermore, the activity of neutral and acid nucleases in skin tissue was determined to be 5.2 and 14.8 U per 1 g of porcine skin tissue, respectively. Investigation of the protective character of different delivery systems revealed that formulations containing DNAzyme in the outer water phase (submicron emulsion and microemulsion) did not exhibit any form of protective effect, whereas formulations containing DNAzyme in the inner water phase (multiple emulsion and W/O emulsion) were able to prevent the DNAzyme degradation to a considerable degree. Consequently, these formulations are promising candidates for the dermal drug delivery of oligonucleotides.


Subject(s)
Chromatography, High Pressure Liquid/methods , DNA, Catalytic/administration & dosage , Drug Delivery Systems , Skin/metabolism , Administration, Cutaneous , Animals , DNA, Catalytic/metabolism , Deoxyribonuclease I/metabolism , Emulsions , Particle Size , Skin/enzymology , Swine , Time Factors
12.
Proc Natl Acad Sci U S A ; 107(47): 20388-93, 2010 Nov 23.
Article in English | MEDLINE | ID: mdl-21059920

ABSTRACT

Mutations of inversin cause type II nephronophthisis, an infantile autosomal recessive disease characterized by cystic kidney disease and developmental defects. Inversin regulates Wnt signaling and is required for convergent extension movements during early embryogenesis. We now show that Inversin is essential for Xenopus pronephros formation, involving two distinct and opposing forms of cell movements. Knockdown of Inversin abrogated both proximal pronephros extension and distal tubule differentiation, phenotypes similar to that of Xenopus deficient in Frizzled-8. Exogenous Inversin rescued the pronephric defects caused by lack of Frizzled-8, indicating that Inversin acts downstream of Frizzled-8 in pronephros morphogenesis. Depletion of Inversin prevents the recruitment of Dishevelled in response to Frizzled-8 and impeded the accumulation of Dishevelled at the apical membrane of tubular epithelial cells in vivo. Thus, defective tubule morphogenesis seems to contribute to the renal pathology observed in patients with nephronophthisis type II.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , Kidney/embryology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Dishevelled Proteins , Fluorescence , In Situ Hybridization , Kidney/metabolism , Mice , Microscopy, Confocal , Oligonucleotides/genetics , Phosphoproteins/metabolism , Wnt Proteins/metabolism , Xenopus
13.
Psychiatry ; 45(2): 115-121, 1982 May.
Article in English | MEDLINE | ID: mdl-27724803
SELECTION OF CITATIONS
SEARCH DETAIL
...