Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 119(18): e2115960119, 2022 05 03.
Article in English | MEDLINE | ID: mdl-35482924

ABSTRACT

Nephronophthisis (NPH) is an autosomal recessive tubulointerstitial nephropathy belonging to the ciliopathy disorders and known as the most common cause of hereditary end-stage renal disease in children. Yet, no curative treatment is available. The major gene, NPHP1, encodes a protein playing key functions at the primary cilium and cellular junctions. Using a medium-throughput drug-screen in NPHP1 knockdown cells, we identified 51 Food and Drug Administration-approved compounds by their ability to alleviate the cellular phenotypes associated with the loss of NPHP1; 11 compounds were further selected for their physicochemical properties. Among those compounds, prostaglandin E1 (PGE1) rescued ciliogenesis defects in immortalized patient NPHP1 urine-derived renal tubular cells, and improved ciliary and kidney phenotypes in our NPH zebrafish and Nphp1 knockout mouse models. Furthermore, Taprenepag, a nonprostanoid prostaglandin E2 receptor agonist, alleviated the severe retinopathy observed in Nphp1−/− mice. Finally, comparative transcriptomics allowed identification of key signaling pathways downstream PGE1, including cell cycle progression, extracellular matrix, adhesion, or actin cytoskeleton organization. In conclusion, using in vitro and in vivo models, we showed that prostaglandin E2 receptor agonists can ameliorate several of the pleotropic phenotypes caused by the absence of NPHP1; this opens their potential as a first therapeutic option for juvenile NPH-associated ciliopathies.


Subject(s)
Ciliopathies , Polycystic Kidney Diseases , Animals , Cilia/metabolism , Ciliopathies/drug therapy , Ciliopathies/genetics , Ciliopathies/metabolism , Female , Humans , Kidney Diseases, Cystic/congenital , Male , Mice , Polycystic Kidney Diseases/metabolism , Prostaglandins/metabolism , Receptors, Prostaglandin E/metabolism , Zebrafish
2.
Hum Mol Genet ; 31(13): 2121-2136, 2022 07 07.
Article in English | MEDLINE | ID: mdl-35043953

ABSTRACT

Renal ciliopathies are the leading cause of inherited kidney failure. In autosomal dominant polycystic kidney disease (ADPKD), mutations in the ciliary gene PKD1 lead to the induction of CCL2, which promotes macrophage infiltration in the kidney. Whether or not mutations in genes involved in other renal ciliopathies also lead to immune cells recruitment is controversial. Through the parallel analysis of patients' derived material and murine models, we investigated the inflammatory components of nephronophthisis (NPH), a rare renal ciliopathy affecting children and adults. Our results show that NPH mutations lead to kidney infiltration by neutrophils, macrophages and T cells. Contrary to ADPKD, this immune cell recruitment does not rely on the induction of CCL2 in mutated cells, which is dispensable for disease progression. Through an unbiased approach, we identified a set of inflammatory cytokines that are upregulated precociously and independently of CCL2 in murine models of NPH. The majority of these transcripts is also upregulated in NPH patient renal cells at a level exceeding those found in common non-immune chronic kidney diseases. This study reveals that inflammation is a central aspect in NPH and delineates a specific set of inflammatory mediators that likely regulates immune cell recruitment in response to NPH genes mutations.


Subject(s)
Ciliopathies , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Adult , Animals , Child , Ciliopathies/genetics , Fibrosis , Humans , Kidney , Mice , Polycystic Kidney, Autosomal Dominant/genetics , TRPP Cation Channels/genetics
3.
Proc Natl Acad Sci U S A ; 117(2): 1113-1118, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31879347

ABSTRACT

Genetic and phenotypic heterogeneity and the lack of sufficiently large patient cohorts pose a significant challenge to understanding genetic associations in rare disease. Here we identify Bsnd (alias Barttin) as a genetic modifier of cystic kidney disease in Joubert syndrome, using a Cep290-deficient mouse model to recapitulate the phenotypic variability observed in patients by mixing genetic backgrounds in a controlled manner and performing genome-wide analysis of these mice. Experimental down-regulation of Bsnd in the parental mouse strain phenocopied the severe cystic kidney phenotype. A common polymorphism within human BSND significantly associates with kidney disease severity in a patient cohort with CEP290 mutations. The striking phenotypic modifications we describe are a timely reminder of the value of mouse models and highlight the significant contribution of genetic background. Furthermore, if appropriately managed, this can be exploited as a powerful tool to elucidate mechanisms underlying human disease heterogeneity.


Subject(s)
Abnormalities, Multiple/genetics , Cerebellum/abnormalities , Chloride Channels/genetics , Chloride Channels/metabolism , Eye Abnormalities/genetics , Genes, Modifier , Kidney Diseases, Cystic/genetics , Retina/abnormalities , Animals , Antigens, Neoplasm/genetics , Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Disease Models, Animal , Genetic Predisposition to Disease/genetics , Kidney Diseases , Mice , Mice, Inbred C57BL , Mutation , Phenotype , Polymorphism, Single Nucleotide , Severity of Illness Index
4.
Am J Hum Genet ; 101(5): 803-814, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100091

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) constitute a major cause of chronic kidney disease in children and 20% of prenatally detected anomalies. CAKUT encompass a spectrum of developmental kidney defects, including renal agenesis, hypoplasia, and cystic and non-cystic dysplasia. More than 50 genes have been reported as mutated in CAKUT-affected case subjects. However, the pathophysiological mechanisms leading to bilateral kidney agenesis (BKA) remain largely elusive. Whole-exome or targeted exome sequencing of 183 unrelated familial and/or severe CAKUT-affected case subjects, including 54 fetuses with BKA, led to the identification of 16 heterozygous variants in GREB1L (growth regulation by estrogen in breast cancer 1-like), a gene reported as a target of retinoic acid signaling. Four loss-of-function and 12 damaging missense variants, 14 being absent from GnomAD, were identified. Twelve of them were present in familial or simplex BKA-affected case subjects. Female BKA-affected fetuses also displayed uterus agenesis. We demonstrated a significant association between GREB1L variants and BKA. By in situ hybridization, we showed expression of Greb1l in the nephrogenic zone in developing mouse kidney. We generated a Greb1l knock-out mouse model by CRISPR-Cas9. Analysis at E13.5 revealed lack of kidneys and genital tract anomalies in male and female Greb1l-/- embryos and a slight decrease in ureteric bud branching in Greb1l+/- embryos. We showed that Greb1l invalidation in mIMCD3 cells affected tubulomorphogenesis in 3D-collagen culture, a phenotype rescued by expression of the wild-type human protein. This demonstrates that GREB1L plays a major role in early metanephros and genital development in mice and humans.


Subject(s)
Congenital Abnormalities/genetics , Kidney Diseases/congenital , Kidney/abnormalities , Mutation/genetics , Neoplasm Proteins/genetics , Proteins/genetics , Animals , Child , Exome/genetics , Female , Fetus/abnormalities , Heterozygote , Humans , Kidney Diseases/genetics , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Urinary Tract/abnormalities , Urogenital Abnormalities/genetics
6.
Am J Hum Genet ; 100(2): 323-333, 2017 02 02.
Article in English | MEDLINE | ID: mdl-28089251

ABSTRACT

Nephronophthisis (NPH), an autosomal-recessive tubulointerstitial nephritis, is the most common cause of hereditary end-stage renal disease in the first three decades of life. Since most NPH gene products (NPHP) function at the primary cilium, NPH is classified as a ciliopathy. We identified mutations in a candidate gene in eight individuals from five families presenting late-onset NPH with massive renal fibrosis. This gene encodes MAPKBP1, a poorly characterized scaffolding protein for JNK signaling. Immunofluorescence analyses showed that MAPKBP1 is not present at the primary cilium and that fibroblasts from affected individuals did not display ciliogenesis defects, indicating that MAPKBP1 may represent a new family of NPHP not involved in cilia-associated functions. Instead, MAPKBP1 is recruited to mitotic spindle poles (MSPs) during the early phases of mitosis where it colocalizes with its paralog WDR62, which plays a key role at MSP. Detected mutations compromise recruitment of MAPKBP1 to the MSP and/or its interaction with JNK2 or WDR62. Additionally, we show increased DNA damage response signaling in fibroblasts from affected individuals and upon knockdown of Mapkbp1 in murine cell lines, a phenotype previously associated with NPH. In conclusion, we identified mutations in MAPKBP1 as a genetic cause of juvenile or late-onset and cilia-independent NPH.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Kidney Diseases, Cystic/congenital , Adolescent , Alleles , Animals , Cell Cycle Proteins , Child , Cilia/genetics , DNA Damage/genetics , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Humans , Kidney/cytology , Kidney/metabolism , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Kidney Failure, Chronic/diagnosis , Kidney Failure, Chronic/genetics , Mice , Mice, Knockout , Mitosis , Mutation , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Pedigree , Phenotype , Signal Transduction , Spindle Poles/metabolism , Young Adult , Zebrafish
7.
PLoS Genet ; 12(3): e1005894, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26967905

ABSTRACT

Ciliopathies are a group of genetic multi-systemic disorders related to dysfunction of the primary cilium, a sensory organelle present at the cell surface that regulates key signaling pathways during development and tissue homeostasis. In order to identify novel genes whose mutations would cause severe developmental ciliopathies, >500 patients/fetuses were analyzed by a targeted high throughput sequencing approach allowing exome sequencing of >1200 ciliary genes. NEK8/NPHP9 mutations were identified in five cases with severe overlapping phenotypes including renal cystic dysplasia/hypodysplasia, situs inversus, cardiopathy with hypertrophic septum and bile duct paucity. These cases highlight a genotype-phenotype correlation, with missense and nonsense mutations associated with hypodysplasia and enlarged cystic organs, respectively. Functional analyses of NEK8 mutations in patient fibroblasts and mIMCD3 cells showed that these mutations differentially affect ciliogenesis, proliferation/apoptosis/DNA damage response, as well as epithelial morphogenesis. Notably, missense mutations exacerbated some of the defects due to NEK8 loss of function, highlighting their likely gain-of-function effect. We also showed that NEK8 missense and loss-of-function mutations differentially affect the regulation of the main Hippo signaling effector, YAP, as well as the expression of its target genes in patient fibroblasts and renal cells. YAP imbalance was also observed in enlarged spheroids of Nek8-invalidated renal epithelial cells grown in 3D culture, as well as in cystic kidneys of Jck mice. Moreover, co-injection of nek8 MO with WT or mutated NEK8-GFP RNA in zebrafish embryos led to shortened dorsally curved body axis, similar to embryos injected with human YAP RNA. Finally, treatment with Verteporfin, an inhibitor of YAP transcriptional activity, partially rescued the 3D spheroid defects of Nek8-invalidated cells and the abnormalities of NEK8-overexpressing zebrafish embryos. Altogether, our study demonstrates that NEK8 human mutations cause major organ developmental defects due to altered ciliogenesis and cell differentiation/proliferation through deregulation of the Hippo pathway.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cilia/genetics , Phosphoproteins/genetics , Polycystic Kidney Diseases/genetics , Protein Kinases/genetics , Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Adaptor Proteins, Signal Transducing/biosynthesis , Animals , Cell Differentiation/genetics , Cilia/pathology , Female , Genetic Association Studies , Humans , Kidney/metabolism , Kidney/pathology , Mice , Morphogenesis/genetics , Mutation , NIMA-Related Kinases , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/biosynthesis , Polycystic Kidney Diseases/pathology , Porphyrins/administration & dosage , Signal Transduction , Transcription Factors , Verteporfin , YAP-Signaling Proteins , Zebrafish
8.
J Am Soc Nephrol ; 25(11): 2435-43, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24876116

ABSTRACT

Several genes, mainly involved in podocyte cytoskeleton regulation, have been implicated in familial forms of primary FSGS. We identified a homozygous missense mutation (p.P209L) in the TTC21B gene in seven families with FSGS. Mutations in this ciliary gene were previously reported to cause nephronophthisis, a chronic tubulointerstitial nephropathy. Notably, tubular basement membrane thickening reminiscent of that observed in nephronophthisis was present in patients with FSGS and the p.P209L mutation. We demonstrated that the TTC21B gene product IFT139, an intraflagellar transport-A component, mainly localizes at the base of the primary cilium in developing podocytes from human fetal tissue and in undifferentiated cultured podocytes. In contrast, in nonciliated adult podocytes and differentiated cultured cells, IFT139 relocalized along the extended microtubule network. We further showed that knockdown of IFT139 in podocytes leads to primary cilia defects, abnormal cell migration, and cytoskeleton alterations, which can be partially rescued by p.P209L overexpression, indicating its hypomorphic effect. Our results demonstrate the involvement of a ciliary gene in a glomerular disorder and point to a critical function of IFT139 in podocytes. Altogether, these data suggest that this homozygous TTC21B p.P209L mutation leads to a novel hereditary kidney disorder with both glomerular and tubulointerstitial damages.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cilia/physiology , Glomerulosclerosis, Focal Segmental/genetics , Microtubule-Associated Proteins/genetics , Podocytes/physiology , Adolescent , Adult , Animals , Cell Line, Transformed , Child , Cilia/pathology , Family Health , Female , Glomerulosclerosis, Focal Segmental/pathology , Haplotypes , Homozygote , Humans , Male , Mice , Mutation, Missense , Pedigree , Phenotype , Podocytes/pathology , Stress Fibers/pathology , Stress Fibers/physiology , Young Adult
9.
Am J Hum Genet ; 94(2): 288-94, 2014 Feb 06.
Article in English | MEDLINE | ID: mdl-24439109

ABSTRACT

Renal hypodysplasia (RHD) is a heterogeneous condition encompassing a spectrum of kidney development defects including renal agenesis, hypoplasia, and (cystic) dysplasia. Heterozygous mutations of several genes have been identified as genetic causes of RHD with various severity. However, these genes and mutations are not associated with bilateral renal agenesis, except for RET mutations, which could be involved in a few cases. The pathophysiological mechanisms leading to total absence of kidney development thus remain largely elusive. By using a whole-exome sequencing approach in families with several fetuses with bilateral renal agenesis, we identified recessive mutations in the integrin α8-encoding gene ITGA8 in two families. Itga8 homozygous knockout in mice is known to result in absence of kidney development. We provide evidence of a damaging effect of the human ITGA8 mutations. These results demonstrate that mutations of ITGA8 are a genetic cause of bilateral renal agenesis and that, at least in some cases, bilateral renal agenesis is an autosomal-recessive disease.


Subject(s)
Congenital Abnormalities/genetics , Genes, Recessive , Integrin alpha Chains/genetics , Kidney Diseases/congenital , Kidney/abnormalities , Urogenital Abnormalities/genetics , Congenital Abnormalities/pathology , Female , Fetus/abnormalities , Homozygote , Humans , Integrin alpha Chains/metabolism , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Male , Mutation , Pedigree , Urogenital Abnormalities/pathology
10.
J Cell Biol ; 198(5): 927-40, 2012 Sep 03.
Article in English | MEDLINE | ID: mdl-22927466

ABSTRACT

Cilia are at the core of planar polarity cellular events in many systems. However, the molecular mechanisms by which they influence the polarization process are unclear. Here, we identify the function of the ciliopathy protein Rpgrip1l in planar polarity. In the mouse cochlea and in the zebrafish floor plate, Rpgrip1l was required for positioning the basal body along the planar polarity axis. Rpgrip1l was also essential for stabilizing dishevelled at the cilium base in the zebrafish floor plate and in mammalian renal cells. In rescue experiments, we showed that in the zebrafish floor plate the function of Rpgrip1l in planar polarity was mediated by dishevelled stabilization. In cultured cells, Rpgrip1l participated in a complex with inversin and nephrocystin-4, two ciliopathy proteins known to target dishevelled to the proteasome, and, in this complex, Rpgrip1l prevented dishevelled degradation. We thus uncover a ciliopathy protein complex that finely tunes dishevelled levels, thereby modulating planar cell polarity processes.


Subject(s)
Cell Polarity/physiology , Cilia/physiology , Proteins/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Line , Cilia/metabolism , Cochlea/metabolism , Cochlea/physiology , Cytoskeletal Proteins , Mice , Mice, Inbred C57BL , Transcription Factors/metabolism , Zebrafish
11.
Hum Mol Genet ; 20(13): 2611-27, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21498478

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-ß-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-ß-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.


Subject(s)
Morphogenesis/genetics , Signal Transduction/genetics , Wnt Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Genetically Modified , Apoptosis/genetics , Cell Line , Cilia/genetics , Cilia/pathology , Dishevelled Proteins , Dogs , HEK293 Cells , Humans , Mitosis/genetics , Phenotype , Phosphoproteins/metabolism , Protein Binding/genetics , Protein Stability , Protein Transport/genetics , Zebrafish Proteins/genetics , beta Catenin/metabolism
12.
J Cell Sci ; 123(Pt 10): 1785-95, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20427320

ABSTRACT

Cilia and flagella are eukaryotic organelles involved in multiple cellular functions. The primary cilium is generally non motile and found in numerous vertebrate cell types where it controls key signalling pathways. Despite a common architecture, ultrastructural data suggest some differences in their organisation. Here, we report the first detailed characterisation of the ciliary pocket, a depression of the plasma membrane in which the primary cilium is rooted. This structure is found at low frequency in kidney epithelial cells (IMCD3) but is associated with virtually all primary cilia in retinal pigment epithelial cells (RPE1). Transmission and scanning electron microscopy, immunofluorescence analysis and videomicroscopy revealed that the ciliary pocket establishes closed links with the actin-based cytoskeleton and that it is enriched in active and dynamic clathrin-coated pits. The existence of the ciliary pocket was confirmed in mouse tissues bearing primary cilia (cumulus), as well as motile cilia and flagella (ependymal cells and spermatids). The ciliary pocket shares striking morphological and functional similarities with the flagellar pocket of Trypanosomatids, a trafficking-specialised membrane domain at the base of the flagellum. Our data therefore highlight the conserved role of membrane trafficking in the vicinity of cilia.


Subject(s)
Actins/metabolism , Cilia/metabolism , Cytoskeleton/metabolism , Endocytosis , Flagella/metabolism , Animals , Cell Line , Cell Movement , Cilia/pathology , Epithelium/pathology , Female , Fibroblasts/pathology , Flagella/pathology , Humans , Membrane Microdomains , Mice , Mice, Inbred C57BL , Ovulation , Zona Pellucida/metabolism
13.
Hum Mol Genet ; 18(24): 4711-23, 2009 Dec 15.
Article in English | MEDLINE | ID: mdl-19755384

ABSTRACT

Nephronophthisis (NPH) is an autosomal recessive disorder characterized by renal fibrosis, tubular basement membrane disruption and corticomedullary cyst formation leading to end-stage renal failure. The disease is caused by mutations in NPHP1-9 genes, which encode the nephrocystins, proteins localized to cell-cell junctions and centrosome/primary cilia. Here, we show that nephrocystin mRNA expression is dramatically increased during cell polarization, and shRNA-mediated knockdown of either NPHP1 or NPHP4 in MDCK cells resulted in delayed tight junction (TJ) formation, abnormal cilia formation and disorganized multi-lumen structures when grown in a three-dimensional collagen matrix. Some of these phenotypes are similar to those reported for cells depleted of the TJ proteins PALS1 or Par3, and interestingly, we demonstrate a physical interaction between these nephrocystins and PALS1 as well as their partners PATJ and Par6 and show their partial co-localization in human renal tubules. Taken together, these results demonstrate that the nephrocystins play an essential role in epithelial cell organization, suggesting a plausible mechanism by which the in vivo histopathologic features of NPH might develop.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Epithelial Cells/physiology , Membrane Proteins/metabolism , Morphogenesis , Nucleoside-Phosphate Kinase/metabolism , src Homology Domains , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Dogs , Epithelial Cells/metabolism , Humans , Membrane Proteins/genetics
14.
Nat Genet ; 39(7): 875-81, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17558409

ABSTRACT

Cerebello-oculo-renal syndrome (CORS), also called Joubert syndrome type B, and Meckel (MKS) syndrome belong to the group of developmental autosomal recessive disorders that are associated with primary cilium dysfunction. Using SNP mapping, we identified missense and truncating mutations in RPGRIP1L (KIAA1005) in both CORS and MKS, and we show that inactivation of the mouse ortholog Rpgrip1l (Ftm) recapitulates the cerebral, renal and hepatic defects of CORS and MKS. In addition, we show that RPGRIP1L colocalizes at the basal body and centrosomes with the protein products of both NPHP6 and NPHP4, known genes associated with MKS, CORS and nephronophthisis (a related renal disorder and ciliopathy). In addition, the RPGRIP1L missense mutations found in CORS individuals diminishes the interaction between RPGRIP1L and nephrocystin-4. Our findings show that mutations in RPGRIP1L can cause the multiorgan phenotypic abnormalities found in CORS or MKS, which therefore represent a continuum of the same underlying disorder.


Subject(s)
Cerebellar Diseases/genetics , Ciliary Motility Disorders/genetics , Encephalocele/genetics , Eye Diseases/genetics , Kidney Diseases/genetics , Proteins/genetics , Animals , Child , Cytoskeletal Proteins , Disease Models, Animal , Humans , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Mutant Strains , Point Mutation , Syndrome
15.
Hum Mol Genet ; 14(5): 645-56, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15661758

ABSTRACT

Nephrocystin and nephrocystin-4 are newly identified proteins involved in familial juvenile nephronophthisis, an autosomal recessive nephropathy characterized by cyst formation and renal fibrosis. Nephrocystin is an adaptor protein that is able to associate with signaling molecules involved in cell adhesion and actin cytoskeleton organization, such as p130Cas, Pyk2, tensin and filamins. Nephrocystin was recently shown to interact and to co-localize with the microtubule component beta-tubulin to the primary cilia in renal epithelial cells, an organelle known to play a key role in the pathogenesis of cystic kidney diseases. In this study, we demonstrated that nephrocystin-4 also localizes to the primary cilia in polarized epithelial tubular cells, particularly at the basal bodies, and associates with microtubule component alpha-tubulin, suggesting a common role for the nephrocystin proteins in ciliary function. However, the co-localization of nephrocystin-4 with the microtubules is not restricted to the primary cilia, as nephrocystin-4 was also detected at the centrosomes of dividing cells and close to the cortical actin cytoskeleton in polarized cells. We also detected p130Cas and Pyk2 in the nephrocystin-4-containing complex, confirming the role of the nephrocystin proteins in cell-cell and cell-matrix adhesion signaling events. Finally, we refined the structural and functional regions involved in the interaction between nephrocystin and nephrocystin-4. These data suggest that nephrocystin and nephrocystin-4 belong to a multifunctional complex localized in actin- and microtubule-based structures involved in cell-cell and cell-matrix adhesion signaling as well as in cell division.


Subject(s)
Centrosome/metabolism , Cilia/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Animals , Cell Line , Cytoskeletal Proteins , Dogs , Humans , Kidney/metabolism , Membrane Proteins , Mutation , Proteins/genetics
16.
Nat Genet ; 32(2): 300-5, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12244321

ABSTRACT

Nephronophthisis, the most common genetic cause of chronic renal failure in children, is a progressive tubulo-interstitial kidney disorder that is inherited as an autosomal recessive trait. The disease is characterized by polyuria, growth retardation and deterioration of renal function during childhood or adolescence. The most prominent histological features are modifications of the tubules with thickening of the basement membrane, interstitial fibrosis and, in the advanced stages, medullary cysts. Nephronophthisis can also be associated with conditions affecting extrarenal organs, such as retinitis pigmentosa (Senior-Løken syndrome) and ocular motor apraxia (Cogan syndrome). Three loci are associated with the juvenile, infantile and adolescent forms, on chromosomes 2q13 (NPHP1; refs 5,6), 9q22 (NPHP2; ref. 7) and 3q21 (NPHP3; ref. 8), respectively. NPHP1, the only gene identified so far, encodes nephrocystin, which contains a Src homology 3 (SH3) domain and interacts with intracytoplasmic proteins involved in cell adhesion. Recently, a second locus associated with the juvenile form of the disease, NPHP4, was mapped to chromosome 1p36 (ref. 14). We carried out haplotype analysis of families affected with nephronophthisis that were not linked to the NPHP1, NPHP2 or NPHP3 loci, using markers covering this region. This allowed us to reduce the NPHP4 interval to a one centimorgan interval between D1S2795 and D1S2870, which contains six genes. We identified five different mutations in one of these genes, designated NPHP4, in unrelated individuals with nephronophthisis. The NPHP4 gene encodes a 1,250-amino acid protein of unknown function that we named nephrocystin-4. We demonstrated the interaction of nephrocystin-4 with nephrocystin suggesting that these two proteins participate in a common signaling pathway.


Subject(s)
Carrier Proteins/genetics , Kidney Diseases, Cystic/genetics , Mutation , Adaptor Proteins, Signal Transducing , Adolescent , Adult , Amino Acid Sequence , Animals , Carrier Proteins/metabolism , Child , Chromosome Mapping , Cytoskeletal Proteins , Female , Humans , Male , Membrane Proteins , Molecular Sequence Data , Pedigree , Protein Binding , Proteins/genetics , Proteins/metabolism , Sequence Alignment , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...