Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 150(3): 533-48, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863007

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exome , Kidney Diseases, Cystic/genetics , Microtubule Proteins/metabolism , Animals , Cilia/metabolism , Gene Knockdown Techniques , Genes, Recessive , Humans , MRE11 Homologue Protein , Mice , Proteins , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
2.
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
3.
Kidney Int ; 105(4): 844-864, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38154558

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) are the predominant cause for chronic kidney disease below age 30 years. Many monogenic forms have been discovered due to comprehensive genetic testing like exome sequencing. However, disease-causing variants in known disease-associated genes only explain a proportion of cases. Here, we aim to unravel underlying molecular mechanisms of syndromic CAKUT in three unrelated multiplex families with presumed autosomal recessive inheritance. Exome sequencing in the index individuals revealed three different rare homozygous variants in FOXD2, encoding a transcription factor not previously implicated in CAKUT in humans: a frameshift in the Arabic and a missense variant each in the Turkish and the Israeli family with segregation patterns consistent with autosomal recessive inheritance. CRISPR/Cas9-derived Foxd2 knockout mice presented with a bilateral dilated kidney pelvis accompanied by atrophy of the kidney papilla and mandibular, ophthalmologic, and behavioral anomalies, recapitulating the human phenotype. In a complementary approach to study pathomechanisms of FOXD2-dysfunction-mediated developmental kidney defects, we generated CRISPR/Cas9-mediated knockout of Foxd2 in ureteric bud-induced mouse metanephric mesenchyme cells. Transcriptomic analyses revealed enrichment of numerous differentially expressed genes important for kidney/urogenital development, including Pax2 and Wnt4 as well as gene expression changes indicating a shift toward a stromal cell identity. Histology of Foxd2 knockout mouse kidneys confirmed increased fibrosis. Further, genome-wide association studies suggest that FOXD2 could play a role for maintenance of podocyte integrity during adulthood. Thus, our studies help in genetic diagnostics of monogenic CAKUT and in understanding of monogenic and multifactorial kidney diseases.


Subject(s)
Embryonic Structures , Forkhead Transcription Factors , Kidney Diseases , Kidney , Nephrons , Urinary Tract , Urogenital Abnormalities , Vesico-Ureteral Reflux , Adult , Animals , Humans , Mice , Genome-Wide Association Study , Kidney/abnormalities , Kidney/embryology , Kidney Diseases/genetics , Mice, Knockout , Nephrons/embryology , Transcription Factors/genetics , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/genetics , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/metabolism
4.
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
5.
BMC Med Inform Decis Mak ; 24(1): 134, 2024 May 24.
Article in English | MEDLINE | ID: mdl-38789985

ABSTRACT

BACKGROUND: There are approximately 8,000 different rare diseases that affect roughly 400 million people worldwide. Many of them suffer from delayed diagnosis. Ciliopathies are rare monogenic disorders characterized by a significant phenotypic and genetic heterogeneity that raises an important challenge for clinical diagnosis. Diagnosis support systems (DSS) applied to electronic health record (EHR) data may help identify undiagnosed patients, which is of paramount importance to improve patients' care. Our objective was to evaluate three online-accessible rare disease DSSs using phenotypes derived from EHRs for the diagnosis of ciliopathies. METHODS: Two datasets of ciliopathy cases, either proven or suspected, and two datasets of controls were used to evaluate the DSSs. Patient phenotypes were automatically extracted from their EHRs and converted to Human Phenotype Ontology terms. We tested the ability of the DSSs to diagnose cases in contrast to controls based on Orphanet ontology. RESULTS: A total of 79 cases and 38 controls were selected. Performances of the DSSs on ciliopathy real world data (best DSS with area under the ROC curve = 0.72) were not as good as published performances on the test set used in the DSS development phase. None of these systems obtained results which could be described as "expert-level". Patients with multisystemic symptoms were generally easier to diagnose than patients with isolated symptoms. Diseases easily confused with ciliopathy generally affected multiple organs and had overlapping phenotypes. Four challenges need to be considered to improve the performances: to make the DSSs interoperable with EHR systems, to validate the performances in real-life settings, to deal with data quality, and to leverage methods and resources for rare and complex diseases. CONCLUSION: Our study provides insights into the complexities of diagnosing highly heterogenous rare diseases and offers lessons derived from evaluation existing DSSs in real-world settings. These insights are not only beneficial for ciliopathy diagnosis but also hold relevance for the enhancement of DSS for various complex rare disorders, by guiding the development of more clinically relevant rare disease DSSs, that could support early diagnosis and finally make more patients eligible for treatment.


Subject(s)
Ciliopathies , Electronic Health Records , Rare Diseases , Humans , Ciliopathies/diagnosis , Rare Diseases/diagnosis , Decision Support Systems, Clinical , Phenotype
6.
Kidney Int ; 104(2): 245-253, 2023 08.
Article in English | MEDLINE | ID: mdl-37244473

ABSTRACT

Nephronophthisis is an autosomal recessive tubulointerstitial nephropathy, belonging to the ciliopathy disorders, characterized by fibrosis and/or cysts. It is the most common genetic cause of kidney failure in children and young adults. Clinically and genetically heterogeneous, it is caused by variants in ciliary genes, resulting in either an isolated kidney disease or syndromic forms in association with other manifestations of ciliopathy disorders. No curative treatment is currently available. Over the past 2 decades, advances in understanding disease mechanisms have identified several dysregulated signaling pathways, some shared with other cystic kidney diseases. Notably, molecules previously developed to target these pathways have shown promising beneficial effects in orthologous mouse models. In addition to these knowledge-based repurposing approaches, unbiased "in cellulo" phenotypic screens of "repurposing" libraries identified small molecules able to rescue the ciliogenesis defects observed in nephronophthisis conditions. Those compounds appeared to act on relevant pathways and, when tested, showed beneficial nephronophthisis-associated kidney and/or extrarenal defects in mice. In this review, we have summarized those studies that highlight the drug repurposing strategies in the context of a rare disorders, such as nephronophthisis-related ciliopathies, with broad genetic heterogeneity and systemic manifestations but with shared disease mechanisms.


Subject(s)
Ciliopathies , Kidney Diseases, Cystic , Polycystic Kidney Diseases , Renal Insufficiency , Animals , Mice , Kidney/pathology , Polycystic Kidney Diseases/genetics , Kidney Diseases, Cystic/drug therapy , Kidney Diseases, Cystic/genetics , Ciliopathies/drug therapy , Ciliopathies/genetics , Renal Insufficiency/complications , Fibrosis , Cilia/pathology
7.
Kidney Int ; 104(2): 378-387, 2023 08.
Article in English | MEDLINE | ID: mdl-37230223

ABSTRACT

Nephronophthisis (NPH) is an autosomal-recessive ciliopathy representing one of the most frequent causes of kidney failure in childhood characterized by a broad clinical and genetic heterogeneity. Applied to one of the worldwide largest cohorts of patients with NPH, genetic analysis encompassing targeted and whole exome sequencing identified disease-causing variants in 600 patients from 496 families with a detection rate of 71%. Of 788 pathogenic variants, 40 known ciliopathy genes were identified. However, the majority of patients (53%) bore biallelic pathogenic variants in NPHP1. NPH-causing gene alterations affected all ciliary modules defined by structural and/or functional subdomains. Seventy six percent of these patients had progressed to kidney failure, of which 18% had an infantile form (under five years) and harbored variants affecting the Inversin compartment or intraflagellar transport complex A. Forty eight percent of patients showed a juvenile (5-15 years) and 34% a late-onset disease (over 15 years), the latter mostly carrying variants belonging to the Transition Zone module. Furthermore, while more than 85% of patients with an infantile form presented with extra-kidney manifestations, it only concerned half of juvenile and late onset cases. Eye involvement represented a predominant feature, followed by cerebellar hypoplasia and other brain abnormalities, liver and skeletal defects. The phenotypic variability was in a large part associated with mutation types, genes and corresponding ciliary modules with hypomorphic variants in ciliary genes playing a role in early steps of ciliogenesis associated with juvenile-to-late onset NPH forms. Thus, our data confirm a considerable proportion of late-onset NPH suggesting an underdiagnosis in adult chronic kidney disease.


Subject(s)
Ciliopathies , Kidney Diseases, Cystic , Kidney Failure, Chronic , Polycystic Kidney Diseases , Adult , Humans , Kidney Failure, Chronic/diagnosis , Polycystic Kidney Diseases/complications , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Mutation , Ciliopathies/genetics
8.
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
9.
Hum Mutat ; 43(3): 347-361, 2022 03.
Article in English | MEDLINE | ID: mdl-35005812

ABSTRACT

We report the screening of a large panel of genes in a series of 100 fetuses (98 families) affected with severe renal defects. Causative variants were identified in 22% of cases, greatly improving genetic counseling. The percentage of variants explaining the phenotype was different according to the type of phenotype. The highest diagnostic yield was found in cases affected with the ciliopathy-like phenotype (11/15 families and, in addition, a single heterozygous or a homozygous Class 3 variant in PKHD1 in three unrelated cases with autosomal recessive polycystic kidney disease). The lowest diagnostic yield was observed in cases with congenital anomalies of the kidney and urinary tract (9/78 families and, in addition, Class 3 variants in GREB1L in three unrelated cases with bilateral renal agenesis). Inheritance was autosomal recessive in nine genes (PKHD1, NPHP3, CEP290, TMEM67, DNAJB11, FRAS1, ACE, AGT, and AGTR1), and autosomal dominant in six genes (PKD1, PKD2, PAX2, EYA1, BICC1, and MYOCD). Finally, we developed an original approach of next-generation sequencing targeted RNA sequencing using the custom capture panel used for the sequencing of DNA, to validate one MYOCD heterozygous splicing variant identified in two male siblings with megabladder and inherited from their healthy mother.


Subject(s)
Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Antigens, Neoplasm , Cell Cycle Proteins/genetics , Cytoskeletal Proteins/genetics , Female , Fetus/abnormalities , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Kidney/abnormalities , Kidney Diseases/congenital , Kidney Diseases/diagnosis , Kidney Diseases/genetics , Male , Mutation , Polycystic Kidney, Autosomal Dominant/genetics
10.
EMBO J ; 37(10)2018 05 15.
Article in English | MEDLINE | ID: mdl-29650680

ABSTRACT

Ciliopathies are life-threatening human diseases caused by defective cilia. They can often be traced back to mutations of genes encoding transition zone (TZ) proteins demonstrating that the understanding of TZ organisation is of paramount importance. The TZ consists of multimeric protein modules that are subject to a stringent assembly hierarchy. Previous reports place Rpgrip1l at the top of the TZ assembly hierarchy in Caenorhabditis elegans By performing quantitative immunofluorescence studies in RPGRIP1L-/- mouse embryos and human embryonic cells, we recognise a different situation in vertebrates in which Rpgrip1l deficiency affects TZ assembly in a cell type-specific manner. In cell types in which the loss of Rpgrip1l alone does not affect all modules, additional truncation or removal of vertebrate-specific Rpgrip1 results in an impairment of all modules. Consequently, Rpgrip1l and Rpgrip1 synergistically ensure the TZ composition in several vertebrate cell types, revealing a higher complexity of TZ assembly in vertebrates than in invertebrates.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/physiology , Cilia/physiology , Embryo, Mammalian/metabolism , Fibroblasts/metabolism , Proteins/physiology , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, Neoplasm , Carrier Proteins/physiology , Cell Cycle Proteins , Cell Membrane Structures , Cells, Cultured , Cytoskeletal Proteins , Embryo, Mammalian/cytology , Fibroblasts/cytology , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/physiology , Transcription Factors/physiology
11.
Kidney Int ; 99(2): 405-409, 2021 02.
Article in English | MEDLINE | ID: mdl-33129895

ABSTRACT

DNAJB11 (DnaJ Heat Shock Protein Family (Hsp40) Member B11) heterozygous loss of function variations have been reported in autosomal dominant cystic kidney disease with extensive fibrosis, associated with maturation and trafficking defect involving both the autosomal dominant polycystic kidney disease protein polycystin-1 and the autosomal dominant tubulointerstitial kidney disease protein uromodulin. Here we show that biallelic pathogenic variations in DNAJB11 lead to a severe fetal disease including enlarged cystic kidneys, dilation and proliferation of pancreatic duct cells, and liver ductal plate malformation, an association known as Ivemark II syndrome. Cysts of the kidney were developed exclusively from uromodulin negative tubular segments. In addition, tubular cells from the affected kidneys had elongated primary cilia, a finding previously reported in ciliopathies. Thus, our data show that the recessive disease associated with DNAJB11 variations is a ciliopathy rather than a disease of the autosomal dominant tubulointerstitial kidney disease spectrum, and prompt screening of DNAJB11 in fetal hyperechogenic/cystic kidneys.


Subject(s)
Abnormalities, Multiple , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , HSP40 Heat-Shock Proteins , Humans , Kidney/abnormalities , Kidney/diagnostic imaging , Liver/abnormalities , Pancreas/abnormalities , Polycystic Kidney, Autosomal Dominant/complications , Polycystic Kidney, Autosomal Dominant/genetics
12.
Hum Mol Genet ; 28(16): 2720-2737, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31042281

ABSTRACT

Mutations in genes encoding components of the intraflagellar transport (IFT) complexes have previously been associated with a spectrum of diseases collectively termed ciliopathies. Ciliopathies relate to defects in the formation or function of the cilium, a sensory or motile organelle present on the surface of most cell types. IFT52 is a key component of the IFT-B complex and ensures the interaction of the two subcomplexes, IFT-B1 and IFT-B2. Here, we report novel IFT52 biallelic mutations in cases with a short-rib thoracic dysplasia (SRTD) or a congenital anomaly of kidney and urinary tract (CAKUT). Combining in vitro and in vivo studies in zebrafish, we showed that SRTD-associated missense mutation impairs IFT-B complex assembly and IFT-B2 ciliary localization, resulting in decreased cilia length. In comparison, CAKUT-associated missense mutation has a mild pathogenicity, thus explaining the lack of skeletal defects in CAKUT case. In parallel, we demonstrated that the previously reported homozygous nonsense IFT52 mutation associated with Sensenbrenner syndrome [Girisha et al. (2016) A homozygous nonsense variant in IFT52 is associated with a human skeletal ciliopathy. Clin. Genet., 90, 536-539] leads to exon skipping and results in a partially functional protein. Finally, our work uncovered a novel role for IFT52 in microtubule network regulation. We showed that IFT52 interacts and partially co-localized with centrin at the distal end of centrioles where it is involved in its recruitment and/or maintenance. Alteration of this function likely contributes to centriole splitting observed in Ift52-/- cells. Altogether, our findings allow a better comprehensive genotype-phenotype correlation among IFT52-related cases and revealed a novel, extra-ciliary role for IFT52, i.e. disruption may contribute to pathophysiological mechanisms.


Subject(s)
Carrier Proteins/genetics , Centrosome/metabolism , Genetic Association Studies , Genetic Predisposition to Disease , Microtubules/metabolism , Mutation , Amino Acid Sequence , Animals , Animals, Genetically Modified , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Child , Child, Preschool , Cilia/metabolism , Consanguinity , DNA Mutational Analysis , Female , Genotype , Homozygote , Humans , Infant , Intracellular Signaling Peptides and Proteins , Male , Pedigree , Phenotype , Protein Binding , Protein Interaction Domains and Motifs/genetics , Trimethoprim, Sulfamethoxazole Drug Combination/metabolism , Exome Sequencing , Zebrafish
13.
Hum Mol Genet ; 28(5): 778-795, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30388224

ABSTRACT

Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.


Subject(s)
Congenital Abnormalities/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Kidney Diseases/congenital , Kidney/abnormalities , Kinesins/genetics , Loss of Function Mutation , Microcephaly/genetics , Oncogene Proteins/genetics , Animals , Congenital Abnormalities/metabolism , Cytokinesis/genetics , Disease Models, Animal , Female , Fluorescent Antibody Technique , Genes, Lethal , Genetic Association Studies/methods , Genetic Loci , Humans , Kidney/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kinesins/chemistry , Kinesins/metabolism , Male , Microcephaly/metabolism , Microcephaly/pathology , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Pedigree , Phenotype , Structure-Activity Relationship , Zebrafish
14.
Pediatr Nephrol ; 36(8): 2361-2369, 2021 08.
Article in English | MEDLINE | ID: mdl-33580824

ABSTRACT

BACKGROUND: Co-occurrence of polycystic kidney disease and hyperinsulinemic hypoglycemia has been reported in children in a few families associated with a variant in the promotor of the PMM2 gene, at position -167 upstream of the coding sequence. PMM2 encodes phosphomannomutase 2, a key enzyme in N-glycosylation. While biallelic coding PMM2 mutations are involved in congenital disorder of glycosylation CDG1A, that particular variant in the promoter of the gene, either in the homozygous state or associated with a mutation in the coding exons of the gene, is thought to restrict the N-glycosylation defect to the kidney and the pancreas. METHODS: Targeted exome sequencing of a panel of genes involved in monogenic kidney diseases. RESULTS: We identified a PMM2 variant at position -167 associated with a pathogenic PMM2 variant in the coding exons in 3 families, comprising 6 cases affected with a cystic kidney disease. The spectrum of phenotypes was very broad, from extremely enlarged fetal cystic kidneys in the context of a COACH-like syndrome, to isolated cystic kidney disease with small kidneys, slowly progressing toward kidney failure in adulthood. Hypoglycemia was reported only in one case. CONCLUSION: These data show that the PMM2 promotor variation, in trans of a PMM2 coding mutation, is associated with a wide spectrum of kidney phenotypes, and is not always associated with extra-renal symptoms. When present, extra-renal defects may include COACH-like syndrome. These data prompt screening of PMM2 in unresolved cases of fetal hyperechogenic/cystic kidneys as well as in cystic kidney disease in children and adults. Graphical Abstract.


Subject(s)
Polycystic Kidney Diseases , Congenital Hyperinsulinism , Humans , Mutation , Phenotype , Phosphotransferases (Phosphomutases) , Promoter Regions, Genetic , Syndrome
15.
Proc Natl Acad Sci U S A ; 115(49): 12489-12494, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30446612

ABSTRACT

Genetic treatments of renal ciliopathies leading to cystic kidney disease would provide a real advance in current therapies. Mutations in CEP290 underlie a ciliopathy called Joubert syndrome (JBTS). Human disease phenotypes include cerebral, retinal, and renal disease, which typically progresses to end stage renal failure (ESRF) within the first two decades of life. While currently incurable, there is often a period of years between diagnosis and ESRF that provides a potential window for therapeutic intervention. By studying patient biopsies, patient-derived kidney cells, and a mouse model, we identify abnormal elongation of primary cilia as a key pathophysiological feature of CEP290-associated JBTS and show that antisense oligonucleotide (ASO)-induced splicing of the mutated exon (41, G1890*) restores protein expression in patient cells. We demonstrate that ASO-induced splicing leading to exon skipping is tolerated, resulting in correct localization of CEP290 protein to the ciliary transition zone, and restoration of normal cilia length in patient kidney cells. Using a gene trap Cep290 mouse model of JBTS, we show that systemic ASO treatment can reduce the cystic burden of diseased kidneys in vivo. These findings indicate that ASO treatment may represent a promising therapeutic approach for kidney disease in CEP290-associated ciliopathy syndromes.


Subject(s)
Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Cerebellum/abnormalities , Exons/genetics , Eye Abnormalities/genetics , Eye Abnormalities/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Nuclear Proteins/genetics , Retina/abnormalities , Adolescent , Animals , Antigens, Neoplasm , Cell Cycle Proteins , Cells, Cultured , Cerebellum/pathology , Cytoskeletal Proteins , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Kidney/cytology , Male , Mice , Mutation , Retina/pathology
16.
Kidney Int ; 98(4): 958-969, 2020 10.
Article in English | MEDLINE | ID: mdl-32505465

ABSTRACT

Biallelic mutations in MAPKBP1 were recently associated with late-onset cilia-independent nephronophthisis. MAPKBP1 was found at mitotic spindle poles but could not be detected at primary cilia or centrosomes. Here, by identification and characterization of novel MAPKBP1 variants, we aimed at further investigating its role in health and disease. Genetic analysis was done by exome sequencing, homozygosity mapping, and a targeted kidney gene panel while coimmunoprecipitation was used to explore wild-type and mutant protein-protein interactions. Expression of MAPKBP1 in non-ciliated HeLa and ciliated inner medullary collecting duct cells enabled co-localization studies by fluorescence microscopy. By next generation sequencing, we identified two novel homozygous MAPKBP1 splice-site variants in patients with nephronophthisis-related chronic kidney disease. Splice-site analyses revealed truncation of C-terminal coiled-coil domains and patient-derived deletion constructs lost their ability to homodimerize and heterodimerize with paralogous WDR62. While wild-type MAPKBP1 exhibited centrosomal, basal body, and microtubule association, mutant proteins lost the latter and showed reduced recruitment to cell cycle dependent centriolar structures. Wild-type and mutant proteins had no reciprocal influence upon co-expression excluding dominant negative effects. Thus, MAPKBP1 appears to be a novel microtubule-binding protein with cell cycle dependent centriolar localization. Truncation of its coiled-coil domain is enough to abrogate its dimerization and results in severely disturbed intracellular localizations. Delineating the impact of impaired dimerization on cell cycle regulation and intracellular kidney signaling may provide new insights into common mechanisms of kidney degeneration. Thus, due to milder clinical presentation, MAPKBP1-associated nephronophthisis should be considered in adult patients with otherwise unexplained chronic kidney disease.


Subject(s)
Centrosome , Polycystic Kidney Diseases , Adult , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Centrosome/metabolism , Cilia/metabolism , Dimerization , Fibrosis , Humans , Intracellular Signaling Peptides and Proteins , Nerve Tissue Proteins/metabolism , Polycystic Kidney Diseases/metabolism
17.
Hum Mol Genet ; 27(2): 266-282, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29121203

ABSTRACT

A child presenting with Mainzer-Saldino syndrome (MZSDS), characterized by renal, retinal and skeletal involvements, was also diagnosed with lung infections and airway ciliary dyskinesia. These manifestations suggested dysfunction of both primary and motile cilia, respectively. Targeted exome sequencing identified biallelic mutations in WDR19, encoding an IFT-A subunit previously associated with MZSDS-related chondrodysplasia, Jeune asphyxiating thoracic dysplasia and cranioectodermal dysplasia, linked to primary cilia dysfunction, and in TEKT1 which encodes tektin-1 an uncharacterized member of the tektin family, mutations of which may cause ciliary dyskinesia. Tektin-1 localizes at the centrosome in cycling cells, at basal bodies of both primary and motile cilia and to the axoneme of motile cilia in airway cells. The identified mutations impaired these localizations. In addition, airway cells from the affected individual showed severe motility defects without major ultrastructural changes. Knockdown of tekt1 in zebrafish resulted in phenotypes consistent with a function for tektin-1 in ciliary motility, which was confirmed by live imaging. Finally, experiments in the zebrafish also revealed a synergistic effect of tekt1 and wdr19. Altogether, our data show genetic interactions between WDR19 and TEKT1 likely contributing to the overall clinical phenotype observed in the affected individual and provide strong evidence for TEKT1 as a new candidate gene for primary ciliary dyskinesia.


Subject(s)
Cilia/genetics , Ciliopathies/genetics , Microtubule Proteins/genetics , Animals , Bone and Bones/abnormalities , Cerebellar Ataxia/genetics , Child , Ciliary Motility Disorders/genetics , Ciliopathies/metabolism , Craniosynostoses/genetics , Cytoskeletal Proteins , Ectodermal Dysplasia/genetics , Exome , Female , Humans , Intracellular Signaling Peptides and Proteins , Microtubule Proteins/metabolism , Mutation , Phenotype , Proteins/genetics , Proteins/metabolism , Retinitis Pigmentosa/genetics , Exome Sequencing , Zebrafish/genetics
18.
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
19.
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
20.
Hum Mol Genet ; 26(23): 4657-4667, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973549

ABSTRACT

Joubert syndrome (JBTS) is the archetypal ciliopathy caused by mutation of genes encoding ciliary proteins leading to multi-system phenotypes, including a cerebello-retinal-renal syndrome. JBTS is genetically heterogeneous, however mutations in CEP290 are a common underlying cause. The renal manifestation of JBTS is a juvenile-onset cystic kidney disease, known as nephronophthisis, typically progressing to end-stage renal failure within the first two decades of life, thus providing a potential window for therapeutic intervention. In order to increase understanding of JBTS and its associated kidney disease and to explore potential treatments, we conducted a comprehensive analysis of primary renal epithelial cells directly isolated from patient urine (human urine-derived renal epithelial cells, hURECs). We demonstrate that hURECs from a JBTS patient with renal disease have elongated and disorganized primary cilia and that this ciliary phenotype is specifically associated with an absence of CEP290 protein. Treatment with the Sonic hedgehog (Shh) pathway agonist purmorphamine or cyclin-dependent kinase inhibition (using roscovitine and siRNA directed towards cyclin-dependent kinase 5) ameliorated the cilia phenotype. In addition, purmorphamine treatment was shown to reduce cyclin-dependent kinase 5 in patient cells, suggesting a convergence of these signalling pathways. To our knowledge, this is the most extensive analysis of primary renal epithelial cells from JBTS patients to date. It demonstrates the feasibility and power of this approach to directly assess the consequences of patient-specific mutations in a physiologically relevant context and a previously unrecognized convergence of Shh agonism and cyclin-dependent kinase inhibition as potential therapeutic targets.


Subject(s)
Abnormalities, Multiple/drug therapy , Abnormalities, Multiple/pathology , Cerebellum/abnormalities , Cilia/pathology , Eye Abnormalities/drug therapy , Eye Abnormalities/pathology , Kidney Diseases, Cystic/drug therapy , Kidney Diseases, Cystic/pathology , Morpholines/therapeutic use , Purines/therapeutic use , Retina/abnormalities , Abnormalities, Multiple/genetics , Abnormalities, Multiple/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Cell Cycle Proteins , Cerebellum/metabolism , Cerebellum/pathology , Child , Child, Preschool , Cilia/drug effects , Cilia/genetics , Cilia/metabolism , Ciliopathies/drug therapy , Ciliopathies/genetics , Ciliopathies/metabolism , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Cytoskeletal Proteins , Epithelial Cells/drug effects , Epithelial Cells/pathology , Eye Abnormalities/genetics , Eye Abnormalities/metabolism , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/metabolism , Kidney Failure, Chronic/pathology , Male , Mutation , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pedigree , Polycystic Kidney Diseases/genetics , Primary Cell Culture , Retina/metabolism , Retina/pathology , Roscovitine , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL