Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Kidney Int ; 104(2): 367-377, 2023 08.
Article in English | MEDLINE | ID: mdl-37230224

ABSTRACT

X-linked Alport syndrome (XLAS) is an inherited kidney disease caused exclusively by pathogenic variants in the COL4A5 gene. In 10-20% of cases, DNA sequencing of COL4A5 exons or flanking regions cannot identify molecular causes. Here, our objective was to use a transcriptomic approach to identify causative events in a group of 19 patients with XLAS without identified mutation by Alport gene panel sequencing. Bulk RNAseq and/or targeted RNAseq using a capture panel of kidney genes was performed. Alternative splicing events were compared to those of 15 controls by a developed bioinformatic score. When using targeted RNAseq, COL4A5 coverage was found to be 23-fold higher than with bulk RNASeq and revealed 30 significant alternative splicing events in 17 of the 19 patients. After computational scoring, a pathogenic transcript was found in all patients. A causative variant affecting COL4A5 splicing and absent in the general population was identified in all cases. Altogether, we developed a simple and robust method for identification of aberrant transcripts due to pathogenic deep-intronic COL4A5 variants. Thus, these variants, potentially targetable by specific antisense oligonucleotide therapies, were found in a high percentage of patients with XLAS in whom pathogenic variants were missed by conventional DNA sequencing.


Subject(s)
Nephritis, Hereditary , Humans , Nephritis, Hereditary/diagnosis , Nephritis, Hereditary/genetics , Nephritis, Hereditary/pathology , Collagen Type IV/genetics , Collagen Type IV/metabolism , Mutation , Exons , RNA Splicing
2.
Clin Genet ; 103(6): 693-698, 2023 06.
Article in English | MEDLINE | ID: mdl-36705481

ABSTRACT

Whole-genome sequencing (WGS) now allows identification of multiple variants in non-coding regions. The large number of variants identified by WGS however complicates their interpretation. Through identification of the first deep intronic variant in NPHS2, which encodes podocin, a protein implicated in autosomal recessive steroid resistant nephrotic syndrome (SRNS), we compare herein three different tools including a newly developed targeted NGS-based RNA-sequencing to explore the splicing effect of intronic variations. WGS identified two different variants in NPHS2 eventually involved in the disease. Through RT-PCR, exon-trapping Minigene assay and targeted RNA sequencing, we were able to identify the splicing defect in NPHS2 mRNA from patient kidney tissue. Only targeted RNA-seq simultaneously analyzed the effect of multiple variants and offered the opportunity to quantify consequences on splicing. Identifying deep intronic variants and their role in disease is of utmost importance. Alternative splicing can be predicted by in silico tools but always requires confirmation through functional testing with RNA analysis from the implicated tissue remaining the gold standard. When several variants with potential effects on splicing are identified by WGS, a targeted RNA sequencing panel could be of great value.


Subject(s)
Nephrotic Syndrome , Humans , Mutation , Whole Genome Sequencing , Nephrotic Syndrome/genetics , RNA, Messenger/genetics
3.
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
4.
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
5.
Proc Natl Acad Sci U S A ; 117(26): 15137-15147, 2020 06 30.
Article in English | MEDLINE | ID: mdl-32554502

ABSTRACT

RNA modifications play a fundamental role in cellular function. Pseudouridylation, the most abundant RNA modification, is catalyzed by the H/ACA small ribonucleoprotein (snoRNP) complex that shares four core proteins, dyskerin (DKC1), NOP10, NHP2, and GAR1. Mutations in DKC1, NOP10, or NHP2 cause dyskeratosis congenita (DC), a disorder characterized by telomere attrition. Here, we report a phenotype comprising nephrotic syndrome, cataracts, sensorineural deafness, enterocolitis, and early lethality in two pedigrees: males with DKC1 p.Glu206Lys and two children with homozygous NOP10 p.Thr16Met. Females with heterozygous DKC1 p.Glu206Lys developed cataracts and sensorineural deafness, but nephrotic syndrome in only one case of skewed X-inactivation. We found telomere attrition in both pedigrees, but no mucocutaneous abnormalities suggestive of DC. Both mutations fall at the dyskerin-NOP10 binding interface in a region distinct from those implicated in DC, impair the dyskerin-NOP10 interaction, and disrupt the catalytic pseudouridylation site. Accordingly, we found reduced pseudouridine levels in the ribosomal RNA (rRNA) of the patients. Zebrafish dkc1 mutants recapitulate the human phenotype and show reduced 18S pseudouridylation, ribosomal dysregulation, and a cell-cycle defect in the absence of telomere attrition. We therefore propose that this human disorder is the consequence of defective snoRNP pseudouridylation and ribosomal dysfunction.


Subject(s)
Cataract/genetics , Cell Cycle Proteins/genetics , Enterocolitis/genetics , Hearing Loss, Sensorineural/genetics , Nephrotic Syndrome/genetics , Nuclear Proteins/genetics , Ribonucleoproteins, Small Nucleolar/genetics , Animals , Child , Female , Genetic Predisposition to Disease , Humans , Longevity , Male , Models, Molecular , Molecular Dynamics Simulation , Mutation , Pedigree , Protein Conformation , RNA, Ribosomal/genetics , Zebrafish
6.
Nat Commun ; 10(1): 3967, 2019 09 03.
Article in English | MEDLINE | ID: mdl-31481669

ABSTRACT

N6-threonyl-carbamoylation of adenosine 37 of ANN-type tRNAs (t6A) is a universal modification essential for translational accuracy and efficiency. The t6A pathway uses two sequentially acting enzymes, YRDC and OSGEP, the latter being a subunit of the multiprotein KEOPS complex. We recently identified mutations in genes encoding four out of the five KEOPS subunits in children with Galloway-Mowat syndrome (GAMOS), a clinically heterogeneous autosomal recessive disease characterized by early-onset steroid-resistant nephrotic syndrome and microcephaly. Here we show that mutations in YRDC cause an extremely severe form of GAMOS whereas mutations in GON7, encoding the fifth KEOPS subunit, lead to a milder form of the disease. The crystal structure of the GON7/LAGE3/OSGEP subcomplex shows that the intrinsically disordered GON7 protein becomes partially structured upon binding to LAGE3. The structure and cellular characterization of GON7 suggest its involvement in the cellular stability and quaternary arrangement of the KEOPS complex.


Subject(s)
Adenosine/analogs & derivatives , GTP-Binding Proteins/genetics , Hernia, Hiatal/genetics , Intrinsically Disordered Proteins/genetics , Microcephaly/genetics , Nephrosis/genetics , Nuclear Proteins/genetics , RNA, Transfer/genetics , RNA-Binding Proteins/genetics , Adenosine/genetics , Child , Female , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/metabolism , Humans , Intrinsically Disordered Proteins/metabolism , Male , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Mutation , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism
8.
PLoS Genet ; 14(5): e1007386, 2018 05.
Article in English | MEDLINE | ID: mdl-29768408

ABSTRACT

Recent evidence suggests that the presence of more than one pathogenic mutation in a single patient is more common than previously anticipated. One of the challenges hereby is to dissect the contribution of each gene mutation, for which animal models such as Drosophila can provide a valuable aid. Here, we identified three families with mutations in ADD3, encoding for adducin-γ, with intellectual disability, microcephaly, cataracts and skeletal defects. In one of the families with additional cardiomyopathy and steroid-resistant nephrotic syndrome (SRNS), we found a homozygous variant in KAT2B, encoding the lysine acetyltransferase 2B, with impact on KAT2B protein levels in patient fibroblasts, suggesting that this second mutation might contribute to the increased disease spectrum. In order to define the contribution of ADD3 and KAT2B mutations for the patient phenotype, we performed functional experiments in the Drosophila model. We found that both mutations were unable to fully rescue the viability of the respective null mutants of the Drosophila homologs, hts and Gcn5, suggesting that they are indeed pathogenic in flies. While the KAT2B/Gcn5 mutation additionally showed a significantly reduced ability to rescue morphological and functional defects of cardiomyocytes and nephrocytes (podocyte-like cells), this was not the case for the ADD3 mutant rescue. Yet, the simultaneous knockdown of KAT2B and ADD3 synergistically impaired kidney and heart function in flies as well as the adhesion and migration capacity of cultured human podocytes, indicating that mutations in both genes may be required for the full clinical manifestation. Altogether, our studies describe the expansion of the phenotypic spectrum in ADD3 deficiency associated with a homozygous likely pathogenic KAT2B variant and thereby identify KAT2B as a susceptibility gene for kidney and heart disease in ADD3-associated disorders.


Subject(s)
Calmodulin-Binding Proteins/genetics , Drosophila/genetics , Mutation , p300-CBP Transcription Factors/genetics , Abnormalities, Multiple/genetics , Adolescent , Adult , Animals , Calmodulin-Binding Proteins/deficiency , Cell Line , Cells, Cultured , DNA Mutational Analysis , Drosophila Proteins/genetics , Female , Heart Diseases/genetics , Homozygote , Humans , Kidney Failure, Chronic/genetics , Male , Pedigree , Phenotype
9.
Biochim Biophys Acta Mol Basis Dis ; 1864(7): 2448-2457, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29660491

ABSTRACT

Interallelic interactions of membrane proteins are not taken into account while evaluating the pathogenicity of sequence variants in autosomal recessive disorders. Podocin, a membrane-anchored component of the slit diaphragm, is encoded by NPHS2, the major gene mutated in hereditary podocytopathies. We formerly showed that its R229Q variant is only pathogenic when trans-associated to specific 3' mutations and suggested the causal role of an abnormal C-terminal dimerization. Here we show by FRET analysis and size exclusion chromatography that podocin oligomerization occurs exclusively through the C-terminal tail (residues 283-382): principally through the first C-terminal helical region (H1, 283-313), which forms a coiled coil as shown by circular dichroism spectroscopy, and through the 332-348 region. We show the principal role of the oligomerization sites in mediating interallelic interactions: while the monomer-forming R286Tfs*17 podocin remains membranous irrespective of the coexpressed podocin variant identity, podocin variants with an intact H1 significantly influence each other's localization (r2 = 0.68, P = 9.2 × 10-32). The dominant negative effect resulting in intracellular retention of the pathogenic F344Lfs*4-R229Q heterooligomer occurs in parallel with a reduction in the FRET efficiency, suggesting the causal role of a conformational rearrangement. On the other hand, oligomerization can also promote the membrane localization: it can prevent the endocytosis of F344Lfs*4 or F344* podocin mutants induced by C-terminal truncation. In conclusion, C-terminal oligomerization of podocin can mediate both a dominant negative effect and interallelic complementation. Interallelic interactions of NPHS2 are not restricted to the R229Q variant and have to be considered in compound heterozygous individuals.


Subject(s)
Intracellular Signaling Peptides and Proteins , Kidney Diseases , Membrane Proteins , Mutation, Missense , Podocytes/metabolism , Protein Multimerization/genetics , Amino Acid Substitution , Cell Line, Transformed , Fluorescence Resonance Energy Transfer , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Diseases/genetics , Kidney Diseases/metabolism , Kidney Diseases/pathology , Membrane Proteins/chemistry , Membrane Proteins/genetics , Membrane Proteins/metabolism , Podocytes/pathology , Protein Domains
10.
J Biol Chem ; 293(11): 4122-4133, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29382718

ABSTRACT

Podocin is a key component of the slit diaphragm in the glomerular filtration barrier, and mutations in the podocin-encoding gene NPHS2 are a common cause of hereditary steroid-resistant nephrotic syndrome. A mutant allele encoding podocin with a p.R138Q amino acid substitution is the most frequent pathogenic variant in European and North American children, and the corresponding mutant protein is poorly expressed and retained in the endoplasmic reticulum both in vitro and in vivo To better understand the defective trafficking and degradation of this mutant, we generated human podocyte cell lines stably expressing podocinwt or podocinR138Q Although it has been proposed that podocin has a hairpin topology, we present evidence for podocinR138QN-glycosylation, suggesting that most of the protein has a transmembrane topology. We find that N-glycosylated podocinR138Q has a longer half-life than non-glycosylated podocinR138Q and that the latter is far more rapidly degraded than podocinwt Consistent with its rapid degradation, podocinR138Q is exclusively degraded by the proteasome, whereas podocinwt is degraded by both the proteasomal and the lysosomal proteolytic machineries. In addition, we demonstrate an enhanced interaction of podocinR138Q with calnexin as the mechanism of endoplasmic reticulum retention. Calnexin knockdown enriches the podocinR138Q non-glycosylated fraction, whereas preventing exit from the calnexin cycle increases the glycosylated fraction. Altogether, we propose a model in which hairpin podocinR138Q is rapidly degraded by the proteasome, whereas transmembrane podocinR138Q degradation is delayed due to entry into the calnexin cycle.


Subject(s)
Calnexin/metabolism , Endoplasmic Reticulum/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Mutant Proteins/metabolism , Mutation , Proteasome Endopeptidase Complex/metabolism , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutant Proteins/genetics , Protein Transport , Proteolysis
11.
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
12.
J Clin Invest ; 127(3): 912-928, 2017 Mar 01.
Article in English | MEDLINE | ID: mdl-28165339

ABSTRACT

Steroid-resistant nephrotic syndrome (SRNS) causes 15% of chronic kidney disease cases. A mutation in 1 of over 40 monogenic genes can be detected in approximately 30% of individuals with SRNS whose symptoms manifest before 25 years of age. However, in many patients, the genetic etiology remains unknown. Here, we have performed whole exome sequencing to identify recessive causes of SRNS. In 7 families with SRNS and facultative ichthyosis, adrenal insufficiency, immunodeficiency, and neurological defects, we identified 9 different recessive mutations in SGPL1, which encodes sphingosine-1-phosphate (S1P) lyase. All mutations resulted in reduced or absent SGPL1 protein and/or enzyme activity. Overexpression of cDNA representing SGPL1 mutations resulted in subcellular mislocalization of SGPL1. Furthermore, expression of WT human SGPL1 rescued growth of SGPL1-deficient dpl1Δ yeast strains, whereas expression of disease-associated variants did not. Immunofluorescence revealed SGPL1 expression in mouse podocytes and mesangial cells. Knockdown of Sgpl1 in rat mesangial cells inhibited cell migration, which was partially rescued by VPC23109, an S1P receptor antagonist. In Drosophila, Sply mutants, which lack SGPL1, displayed a phenotype reminiscent of nephrotic syndrome in nephrocytes. WT Sply, but not the disease-associated variants, rescued this phenotype. Together, these results indicate that SGPL1 mutations cause a syndromic form of SRNS.


Subject(s)
Aldehyde-Lyases , Cell Movement/genetics , Ichthyosis, Lamellar , Mesangial Cells/enzymology , Mutation , Nephrotic Syndrome , Aldehyde-Lyases/genetics , Aldehyde-Lyases/metabolism , Animals , Cell Line , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster , Female , Humans , Ichthyosis, Lamellar/enzymology , Ichthyosis, Lamellar/genetics , Ichthyosis, Lamellar/pathology , Male , Mesangial Cells/pathology , Mice , Mice, Knockout , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/genetics , Nephrotic Syndrome/pathology , Protein Transport/genetics , Rats
13.
Am J Hum Genet ; 95(6): 637-48, 2014 Dec 04.
Article in English | MEDLINE | ID: mdl-25466283

ABSTRACT

Galloway-Mowat syndrome is a rare autosomal-recessive condition characterized by nephrotic syndrome associated with microcephaly and neurological impairment. Through a combination of autozygosity mapping and whole-exome sequencing, we identified WDR73 as a gene in which mutations cause Galloway-Mowat syndrome in two unrelated families. WDR73 encodes a WD40-repeat-containing protein of unknown function. Here, we show that WDR73 was present in the brain and kidney and was located diffusely in the cytoplasm during interphase but relocalized to spindle poles and astral microtubules during mitosis. Fibroblasts from one affected child and WDR73-depleted podocytes displayed abnormal nuclear morphology, low cell viability, and alterations of the microtubule network. These data suggest that WDR73 plays a crucial role in the maintenance of cell architecture and cell survival. Altogether, WDR73 mutations cause Galloway-Mowat syndrome in a particular subset of individuals presenting with late-onset nephrotic syndrome, postnatal microcephaly, severe intellectual disability, and homogenous brain MRI features. WDR73 is another example of a gene involved in a disease affecting both the kidney glomerulus and the CNS.


Subject(s)
Hernia, Hiatal/genetics , Intellectual Disability/genetics , Microcephaly/genetics , Nephrosis/genetics , Nephrotic Syndrome/genetics , Proteins/genetics , Adolescent , Brain/physiopathology , Cell Line , Cell Survival , Child , Child, Preschool , Cytosol/metabolism , Exome/genetics , Hernia, Hiatal/physiopathology , Homozygote , Humans , Kidney Glomerulus/physiopathology , Male , Microcephaly/physiopathology , Microtubules/metabolism , Mitosis , Models, Molecular , Mutation , Nephrosis/physiopathology , Nephrotic Syndrome/physiopathology , Podocytes , Protein Transport , Proteins/metabolism , Spindle Poles/metabolism
14.
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
15.
Nat Genet ; 46(3): 299-304, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24509478

ABSTRACT

Monogenic disorders result from defects in a single gene. According to Mendel's laws, these disorders are inherited in either a recessive or dominant fashion. Autosomal-recessive disorders require a disease-causing variant on both alleles, and according to our current understanding, their pathogenicities are not influenced by each other. Here we present an autosomal-recessive disorder, nephrotic syndrome type 2 (MIM 600995), in which the pathogenicity of an NPHS2 allele encoding p.Arg229Gln depends on the trans-associated mutation. We show that, contrary to expectations, this allele leads to a disease phenotype only when it is associated specifically with certain 3' NPHS2 mutations because of an altered heterodimerization and mislocalization of the encoded p.Arg229Gln podocin. The disease-associated 3' mutations exert a dominant-negative effect on p.Arg229Gln podocin but behave as recessive alleles when associated with wild-type podocin. Therefore, the transmission rates for couples carrying the disease-associated mutations and p.Arg229Gln may be substantially different from those expected in autosomal-recessive disorders.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Nephrotic Syndrome/congenital , Adult , Amino Acid Substitution , Cell Membrane/metabolism , Child , Cohort Studies , Exons , Female , Gene Frequency , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Models, Molecular , Nephrotic Syndrome/genetics , Nephrotic Syndrome/metabolism , Nephrotic Syndrome/pathology , Podocytes/metabolism , Podocytes/pathology , Protein Multimerization , Protein Structure, Quaternary
16.
Pediatr Nephrol ; 29(7): 1221-30, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24477978

ABSTRACT

BACKGROUND: Fetuses exposed to angiotensin-converting enzyme inhibitors or angiotensin receptor antagonists during the second and/or third trimesters of gestation are at high risk of developing severe complications. They consist in fetal hypotension, and anuria/oligohydramnios leading to Potter sequence, frequently associated with hypocalvaria. Most fetuses die during the pre- or postnatal period, whereas others recover normal or subnormal renal function. However, the secondary occurrence of renal failure or hypertension has been reported in children after apparent complete recovery. METHODS: In this context, we analyzed renal lesions in 14 fetus/neonates who died soon after exposure to renin-angiotensin-system (RAS) blockers. Our objective was to determine the causes for the persistence or the secondary occurrence of renal complications reported in some of the survivors. RESULTS: As previously described, renal tubular dysgenesis is usually observed. Additional lesions, such as thickening of the muscular wall of arterioles and interlobular arteries, glomerular cysts, and interstitial fibrosis, develop early during fetal life. CONCLUSION: We suggest that renal lesions that develop before birth may persist after withdrawal of the causative drugs and normalization of blood and renal perfusion pressure. Their persistence could explain the severe long-term outcome of some of these patients. Long-term study of children exposed to RAS blockers during fetal life is strongly recommended.


Subject(s)
Angiotensin Receptor Antagonists/adverse effects , Angiotensin-Converting Enzyme Inhibitors/adverse effects , Fetal Diseases/chemically induced , Kidney Tubules/abnormalities , Kidney/drug effects , Abnormalities, Drug-Induced , Humans , Immunohistochemistry , Infant, Newborn , Kidney/chemistry , Kidney/pathology , Renin/analysis
17.
Kidney Int ; 83(6): 1052-64, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23364521

ABSTRACT

A thorough characterization of the transcriptome and proteome of endogenous podocytes has been hampered by low cell yields during isolation. Here we describe a double fluorescent reporter mouse model combined with an optimized bead perfusion protocol and efficient single cell dissociation to yield more than 500,000 podocytes per mouse allowing for global, unbiased downstream applications. Combining mRNA and miRNA transcriptional profiling with quantitative proteomic analyses revealed programs of highly specific gene regulation tightly controlling cytoskeleton, cell differentiation, endosomal transport, and peroxisome function in podocytes. Strikingly, the analyses further predict that these podocyte-specific gene regulatory networks are accompanied by alternative splicing of respective genes. Thus, our 'omics' approach will facilitate the discovery and integration of novel gene, protein, and organelle regulatory networks that deepen our systematic understanding of podocyte biology.


Subject(s)
Gene Expression Profiling , Gene Regulatory Networks , Luminescent Proteins/biosynthesis , Podocytes/metabolism , Proteomics , Signal Transduction , Alternative Splicing , Animals , Cell Separation , Computational Biology , Gene Expression Profiling/methods , Genes, Reporter , Genotype , Luminescent Proteins/genetics , Mass Spectrometry , Mice , Mice, Transgenic , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Phenotype , Proteomics/methods , Signal Transduction/genetics
18.
Hum Mutat ; 33(2): 316-26, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22095942

ABSTRACT

Autosomal recessive renal tubular dysgenesis (RTD) is a severe disorder of renal tubular development characterized by early onset and persistent fetal anuria leading to oligohydramnios and the Potter sequence, associated with skull ossification defects. Early death occurs in most cases from anuria, pulmonary hypoplasia, and refractory arterial hypotension. The disease is linked to mutations in the genes encoding several components of the renin-angiotensin system (RAS): AGT (angiotensinogen), REN (renin), ACE (angiotensin-converting enzyme), and AGTR1 (angiotensin II receptor type 1). Here, we review the series of 54 distinct mutations identified in 48 unrelated families. Most of them are novel and ACE mutations are the most frequent, observed in two-thirds of families (64.6%). The severity of the clinical course was similar whatever the mutated gene, which underlines the importance of a functional RAS in the maintenance of blood pressure and renal blood flow during the life of a human fetus. Renal hypoperfusion, whether genetic or secondary to a variety of diseases, precludes the normal development/ differentiation of proximal tubules. The identification of the disease on the basis of precise clinical and histological analyses and the characterization of the genetic defects allow genetic counseling and early prenatal diagnosis.


Subject(s)
Genes, Recessive , Mutation , Renin-Angiotensin System/genetics , Urogenital Abnormalities/genetics , Angiotensinogen/genetics , Animals , Disease Models, Animal , Genetic Association Studies , Humans , Kidney Tubules, Proximal/abnormalities , Peptidyl-Dipeptidase A/genetics , Receptor, Angiotensin, Type 1/genetics , Renin/genetics , Urogenital Abnormalities/diagnosis
19.
N Engl J Med ; 365(25): 2377-88, 2011 Dec 22.
Article in English | MEDLINE | ID: mdl-22187985

ABSTRACT

BACKGROUND: Charcot-Marie-Tooth neuropathy has been reported to be associated with renal diseases, mostly focal segmental glomerulosclerosis (FSGS). However, the common mechanisms underlying the neuropathy and FSGS remain unknown. Mutations in INF2 were recently identified in patients with autosomal dominant FSGS. INF2 encodes a formin protein that interacts with the Rho-GTPase CDC42 and myelin and lymphocyte protein (MAL) that are implicated in essential steps of myelination and myelin maintenance. We therefore hypothesized that INF2 may be responsible for cases of Charcot-Marie-Tooth neuropathy associated with FSGS. METHODS: We performed direct genotyping of INF2 in 16 index patients with Charcot-Marie-Tooth neuropathy and FSGS who did not have a mutation in PMP22 or MPZ, encoding peripheral myelin protein 22 and myelin protein zero, respectively. Histologic and functional studies were also conducted. RESULTS: We identified nine new heterozygous mutations in 12 of the 16 index patients (75%), all located in exons 2 and 3, encoding the diaphanous-inhibitory domain of INF2. Patients presented with an intermediate form of Charcot-Marie-Tooth neuropathy as well as a glomerulopathy with FSGS on kidney biopsy. Immunohistochemical analysis revealed strong INF2 expression in Schwann-cell cytoplasm and podocytes. Moreover, we demonstrated that INF2 colocalizes and interacts with MAL in Schwann cells. The INF2 mutants perturbed the INF2-MAL-CDC42 pathway, resulting in cytoskeleton disorganization, enhanced INF2 binding to CDC42 and mislocalization of INF2, MAL, and CDC42. CONCLUSIONS: INF2 mutations appear to cause many cases of FSGS-associated Charcot-Marie-Tooth neuropathy, showing that INF2 is involved in a disease affecting both the kidney glomerulus and the peripheral nervous system. These findings provide new insights into the pathophysiological mechanisms linking formin proteins to podocyte and Schwann-cell function. (Funded by the Agence Nationale de la Recherche and others.).


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Glomerulosclerosis, Focal Segmental/etiology , Kidney/metabolism , Microfilament Proteins/genetics , Schwann Cells/metabolism , Actins/metabolism , Adolescent , Adult , Age of Onset , Animals , Charcot-Marie-Tooth Disease/complications , Child , Female , Formins , Heterozygote , Humans , Male , Membrane Transport Proteins/metabolism , Mice , Microfilament Proteins/metabolism , Middle Aged , Mutation , Myelin Proteins/metabolism , Myelin and Lymphocyte-Associated Proteolipid Proteins , Phenotype , Proteolipids/metabolism , Young Adult
20.
Hum Mol Genet ; 19(1): 1-15, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19797313

ABSTRACT

The Wilms tumor-suppressor gene WT1, a key player in renal development, also has a crucial role in maintenance of the glomerulus in the mature kidney. However, molecular pathways orchestrated by WT1 in podocytes, where it is highly expressed, remain unknown. Their defects are thought to modify the cross-talk between podocytes and other glomerular cells and ultimately lead to glomerular sclerosis, as observed in diffuse mesangial sclerosis (DMS) a nephropathy associated with WT1 mutations. To identify podocyte WT1 targets, we generated a novel DMS mouse line, performed gene expression profiling in isolated glomeruli and identified excellent candidates that may modify podocyte differentiation and growth factor signaling in glomeruli. Scel, encoding sciellin, a protein of the cornified envelope in the skin, and Sulf1, encoding a 6-O endosulfatase, are shown to be expressed in wild-type podocytes and to be strongly down-regulated in mutants. Co-expression of Wt1, Scel and Sulf1 was also found in a mesonephric cell line, and siRNA-mediated knockdown of WT1 decreased Scel and Sulf1 mRNAs and proteins. By ChIP we show that Scel and Sulf1 are direct WT1 targets. Cyp26a1, encoding an enzyme involved in the degradation of retinoic acid, is shown to be up-regulated in mutant podocytes. Cyp26a1 may play a role in the development of glomerular lesions but does not seem to be regulated by WT1. These results provide novel clues in our understanding of normal glomerular function and early events involved in glomerulosclerosis.


Subject(s)
Denys-Drash Syndrome/genetics , Podocytes/metabolism , Transcription, Genetic , WT1 Proteins/metabolism , Alleles , Animals , Cell Line , Cytochrome P-450 Enzyme System/metabolism , Denys-Drash Syndrome/enzymology , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation , Heterozygote , Kidney Glomerulus/enzymology , Kidney Glomerulus/pathology , Mesonephros/enzymology , Mice , Mice, Inbred Strains , Mutation/genetics , Protein Binding , Protein Transport , RNA Transport , Regulatory Sequences, Nucleic Acid/genetics , Retinoic Acid 4-Hydroxylase , Sulfotransferases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...