Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
2.
J Am Soc Nephrol ; 34(2): 273-290, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36414417

RÉSUMÉ

BACKGROUND: About 40 disease genes have been described to date for isolated CAKUT, the most common cause of childhood CKD. However, these genes account for only 20% of cases. ARHGEF6, a guanine nucleotide exchange factor that is implicated in biologic processes such as cell migration and focal adhesion, acts downstream of integrin-linked kinase (ILK) and parvin proteins. A genetic variant of ILK that causes murine renal agenesis abrogates the interaction of ILK with a murine focal adhesion protein encoded by Parva , leading to CAKUT in mice with this variant. METHODS: To identify novel genes that, when mutated, result in CAKUT, we performed exome sequencing in an international cohort of 1265 families with CAKUT. We also assessed the effects in vitro of wild-type and mutant ARHGEF6 proteins, and the effects of Arhgef6 deficiency in mouse and frog models. RESULTS: We detected six different hemizygous variants in the gene ARHGEF6 (which is located on the X chromosome in humans) in eight individuals from six families with CAKUT. In kidney cells, overexpression of wild-type ARHGEF6 -but not proband-derived mutant ARHGEF6 -increased active levels of CDC42/RAC1, induced lamellipodia formation, and stimulated PARVA-dependent cell spreading. ARHGEF6-mutant proteins showed loss of interaction with PARVA. Three-dimensional Madin-Darby canine kidney cell cultures expressing ARHGEF6-mutant proteins exhibited reduced lumen formation and polarity defects. Arhgef6 deficiency in mouse and frog models recapitulated features of human CAKUT. CONCLUSIONS: Deleterious variants in ARHGEF6 may cause dysregulation of integrin-parvin-RAC1/CDC42 signaling, thereby leading to X-linked CAKUT.


Sujet(s)
Voies urinaires , Malformations urogénitales , Humains , Souris , Animaux , Chiens , Malformations urogénitales/génétique , Rein/malformations , Voies urinaires/malformations , Intégrines/métabolisme , Protéines mutantes/métabolisme , Rho guanine nucleotide exchange factors/génétique
3.
Genet Med ; 24(2): 307-318, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34906515

RÉSUMÉ

PURPOSE: Congenital anomalies of the kidneys and urinary tract (CAKUT) constitute the leading cause of chronic kidney disease in children. In total, 174 monogenic causes of isolated or syndromic CAKUT are known. However, syndromic features may be overlooked when the initial clinical diagnosis of CAKUT is made. We hypothesized that the yield of a molecular genetic diagnosis by exome sequencing (ES) can be increased by applying reverse phenotyping, by re-examining the case for signs/symptoms of the suspected clinical syndrome that results from the genetic variant detected by ES. METHODS: We conducted ES in an international cohort of 731 unrelated families with CAKUT. We evaluated ES data for variants in 174 genes, in which variants are known to cause isolated or syndromic CAKUT. In cases in which ES suggested a previously unreported syndromic phenotype, we conducted reverse phenotyping. RESULTS: In 83 of 731 (11.4%) families, we detected a likely CAKUT-causing genetic variant consistent with an isolated or syndromic CAKUT phenotype. In 19 of these 83 families (22.9%), reverse phenotyping yielded syndromic clinical findings, thereby strengthening the genotype-phenotype correlation. CONCLUSION: We conclude that employing reverse phenotyping in the evaluation of syndromic CAKUT genes by ES provides an important tool to facilitate molecular genetic diagnostics in CAKUT.


Sujet(s)
Voies urinaires , Malformations urogénitales , Allèles , Exome/génétique , Humains , Rein/malformations , Malformations urogénitales/génétique , Reflux vésico-urétéral
5.
J Allergy Clin Immunol ; 148(2): 381-393, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33872655

RÉSUMÉ

BACKGROUND: Recognition of viral nucleic acids is one of the primary triggers for a type I interferon-mediated antiviral immune response. Inborn errors of type I interferon immunity can be associated with increased inflammation and/or increased susceptibility to viral infections as a result of dysbalanced interferon production. NFX1-type zinc finger-containing 1 (ZNFX1) is an interferon-stimulated double-stranded RNA sensor that restricts the replication of RNA viruses in mice. The role of ZNFX1 in the human immune response is not known. OBJECTIVE: We studied 15 patients from 8 families with an autosomal recessive immunodeficiency characterized by severe infections by both RNA and DNA viruses and virally triggered inflammatory episodes with hemophagocytic lymphohistiocytosis-like disease, early-onset seizures, and renal and lung disease. METHODS: Whole exome sequencing was performed on 13 patients from 8 families. We investigated the transcriptome, posttranscriptional regulation of interferon-stimulated genes (ISGs) and predisposition to viral infections in primary cells from patients and controls stimulated with synthetic double-stranded nucleic acids. RESULTS: Deleterious homozygous and compound heterozygous ZNFX1 variants were identified in all 13 patients. Stimulation of patient-derived primary cells with synthetic double-stranded nucleic acids was associated with a deregulated pattern of expression of ISGs and alterations in the half-life of the mRNA of ISGs and also associated with poorer clearance of viral infections by monocytes. CONCLUSION: ZNFX1 is an important regulator of the response to double-stranded nucleic acids stimuli following viral infections. ZNFX1 deficiency predisposes to severe viral infections and a multisystem inflammatory disease.


Sujet(s)
Antigènes néoplasiques/génétique , , Prédisposition génétique à une maladie , Maladies d'immunodéficience primaire/immunologie , Maladies virales/génétique , Antigènes néoplasiques/immunologie , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Inflammation/imagerie diagnostique , Inflammation/génétique , Inflammation/immunologie , Mâle , Maladies d'immunodéficience primaire/imagerie diagnostique , Maladies d'immunodéficience primaire/génétique , Maladies virales/imagerie diagnostique , Maladies virales/immunologie
6.
Sci Adv ; 7(1)2021 01.
Article de Anglais | MEDLINE | ID: mdl-33523862

RÉSUMÉ

Nephrotic syndrome (NS) is a leading cause of chronic kidney disease. We found recessive NOS1AP variants in two families with early-onset NS by exome sequencing. Overexpression of wild-type (WT) NOS1AP, but not cDNA constructs bearing patient variants, increased active CDC42 and promoted filopodia and podosome formation. Pharmacologic inhibition of CDC42 or its effectors, formin proteins, reduced NOS1AP-induced filopodia formation. NOS1AP knockdown reduced podocyte migration rate (PMR), which was rescued by overexpression of WT Nos1ap but not by constructs bearing patient variants. PMR in NOS1AP knockdown podocytes was also rescued by constitutively active CDC42Q61L or the formin DIAPH3 Modeling a NOS1AP patient variant in knock-in human kidney organoids revealed malformed glomeruli with increased apoptosis. Nos1apEx3-/Ex3- mice recapitulated the human phenotype, exhibiting proteinuria, foot process effacement, and glomerulosclerosis. These findings demonstrate that recessive NOS1AP variants impair CDC42/DIAPH-dependent actin remodeling, cause aberrant organoid glomerulogenesis, and lead to a glomerulopathy in humans and mice.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Maladies du rein , Syndrome néphrotique , Podocytes , Actines/génétique , Actines/métabolisme , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Formines/génétique , Humains , Maladies du rein/métabolisme , Souris , Syndrome néphrotique/génétique , Syndrome néphrotique/métabolisme , Podocytes/métabolisme
7.
Kidney Int Rep ; 6(2): 460-471, 2021 Feb.
Article de Anglais | MEDLINE | ID: mdl-33615071

RÉSUMÉ

INTRODUCTION: Steroid-resistant nephrotic syndrome (SRNS) is the second most common cause of chronic kidney disease during childhood. Identification of 63 monogenic human genes has delineated 12 distinct pathogenic pathways. METHODS: Here, we generated 2 independent sets of nephrotic syndrome (NS) candidate genes to augment the discovery of additional monogenic causes based on whole-exome sequencing (WES) data from 1382 families with NS. RESULTS: We first identified 63 known monogenic causes of NS in mice from public databases and scientific publications, and 12 of these genes overlapped with the 63 known human monogenic SRNS genes. Second, we used a set of 64 genes that are regulated by the transcription factor Wilms tumor 1 (WT1), which causes SRNS if mutated. Thirteen of these WT1-regulated genes overlapped with human or murine NS genes. Finally, we overlapped these lists of murine and WT1 candidate genes with our list of 120 candidate genes generated from WES in 1382 NS families, to identify novel candidate genes for monogenic human SRNS. Using this approach, we identified 7 overlapping genes, of which 3 genes were shared by all datasets, including SYNPO. We show that loss-of-function of SYNPO leads to decreased CDC42 activity and reduced podocyte migration rate, both of which are rescued by overexpression of wild-type complementary DNA (cDNA), but not by cDNA representing the patient mutation. CONCLUSION: Thus, we identified 3 novel candidate genes for human SRNS using 3 independent, nonoverlapping hypotheses, and generated functional evidence for SYNPO as a novel potential monogenic cause of NS.

8.
Kidney Int Rep ; 6(2): 472-483, 2021 Feb.
Article de Anglais | MEDLINE | ID: mdl-33615072

RÉSUMÉ

INTRODUCTION: Most of the approximately 60 genes that if mutated cause steroid-resistant nephrotic syndrome (SRNS) are highly expressed in the glomerular podocyte, rendering SRNS a "podocytopathy." METHODS: We performed whole-exome sequencing (WES) in 1200 nephrotic syndrome (NS) patients. RESULTS: We discovered homozygous truncating and homozygous missense mutation in SYNPO2 (synaptopodin-2) (p.Lys1124∗ and p.Ala1134Thr) in 2 patients with childhood-onset NS. We found SYNPO2 expression in both podocytes and mesangial cells; however, notably, immunofluorescence staining of adult human and rat kidney cryosections indicated that SYNPO2 is localized mainly in mesangial cells. Subcellular localization studies reveal that in these cells SYNPO2 partially co-localizes with α-actinin and filamin A-containing F-actin filaments. Upon transfection in mesangial cells or podocytes, EGFP-SYNPO2 co-localized with α-actinin-4, which gene is mutated in autosomal dominant SRNS in humans. SYNPO2 overexpression increases mesangial cell migration rate (MMR), whereas shRNA knockdown reduces MMR. Decreased MMR was rescued by transfection of wild-type mouse Synpo2 cDNA but only partially by cDNA representing mutations from the NS patients. The increased mesangial cell migration rate (MMR) by SYNPO2 overexpression was inhibited by ARP complex inhibitor CK666. SYNPO2 shRNA knockdown in podocytes decreased active Rac1, which was rescued by transfection of wild-type SYNPO2 cDNA but not by cDNA representing any of the 2 mutant variants. CONCLUSION: We show that SYNPO2 variants may lead to Rac1-ARP3 dysregulation, and may play a role in the pathogenesis of nephrotic syndrome.

9.
J Am Soc Nephrol ; 32(3): 580-596, 2021 03.
Article de Anglais | MEDLINE | ID: mdl-33593823

RÉSUMÉ

BACKGROUND: Galloway-Mowat syndrome (GAMOS) is characterized by neurodevelopmental defects and a progressive nephropathy, which typically manifests as steroid-resistant nephrotic syndrome. The prognosis of GAMOS is poor, and the majority of children progress to renal failure. The discovery of monogenic causes of GAMOS has uncovered molecular pathways involved in the pathogenesis of disease. METHODS: Homozygosity mapping, whole-exome sequencing, and linkage analysis were used to identify mutations in four families with a GAMOS-like phenotype, and high-throughput PCR technology was applied to 91 individuals with GAMOS and 816 individuals with isolated nephrotic syndrome. In vitro and in vivo studies determined the functional significance of the mutations identified. RESULTS: Three biallelic variants of the transcriptional regulator PRDM15 were detected in six families with proteinuric kidney disease. Four families with a variant in the protein's zinc-finger (ZNF) domain have additional GAMOS-like features, including brain anomalies, cardiac defects, and skeletal defects. All variants destabilize the PRDM15 protein, and the ZNF variant additionally interferes with transcriptional activation. Morpholino oligonucleotide-mediated knockdown of Prdm15 in Xenopus embryos disrupted pronephric development. Human wild-type PRDM15 RNA rescued the disruption, but the three PRDM15 variants did not. Finally, CRISPR-mediated knockout of PRDM15 in human podocytes led to dysregulation of several renal developmental genes. CONCLUSIONS: Variants in PRDM15 can cause either isolated nephrotic syndrome or a GAMOS-type syndrome on an allelic basis. PRDM15 regulates multiple developmental kidney genes, and is likely to play an essential role in renal development in humans.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Hernie hiatale/génétique , Microcéphalie/génétique , Mutation faux-sens , Néphrose/génétique , Facteurs de transcription/génétique , Séquence d'acides aminés , Substitution d'acide aminé , Animaux , Lignée cellulaire , Enfant d'âge préscolaire , Protéines de liaison à l'ADN/composition chimique , Protéines de liaison à l'ADN/déficit , Femelle , Régulation de l'expression des gènes au cours du développement , Techniques de knock-down de gènes , Techniques de knock-out de gènes , Séquençage nucléotidique à haut débit , Humains , Nourrisson , Nouveau-né , Mâle , Modèles moléculaires , Syndrome néphrotique/génétique , Podocytes/métabolisme , Polymorphisme de nucléotide simple , Pronéphros/embryologie , Pronéphros/métabolisme , Stabilité protéique , Facteurs de transcription/composition chimique , Facteurs de transcription/déficit , Xenopus laevis/embryologie , Xenopus laevis/génétique , Doigts de zinc/génétique
10.
Am J Hum Genet ; 108(2): 357-367, 2021 02 04.
Article de Anglais | MEDLINE | ID: mdl-33508234

RÉSUMÉ

Focal segmental glomerulosclerosis (FSGS) is the main pathology underlying steroid-resistant nephrotic syndrome (SRNS) and a leading cause of chronic kidney disease. Monogenic forms of pediatric SRNS are predominantly caused by recessive mutations, while the contribution of de novo variants (DNVs) to this trait is poorly understood. Using exome sequencing (ES) in a proband with FSGS/SRNS, developmental delay, and epilepsy, we discovered a nonsense DNV in TRIM8, which encodes the E3 ubiquitin ligase tripartite motif containing 8. To establish whether TRIM8 variants represent a cause of FSGS, we aggregated exome/genome-sequencing data for 2,501 pediatric FSGS/SRNS-affected individuals and 48,556 control subjects, detecting eight heterozygous TRIM8 truncating variants in affected subjects but none in control subjects (p = 3.28 × 10-11). In all six cases with available parental DNA, we demonstrated de novo inheritance (p = 2.21 × 10-15). Reverse phenotyping revealed neurodevelopmental disease in all eight families. We next analyzed ES from 9,067 individuals with epilepsy, yielding three additional families with truncating TRIM8 variants. Clinical review revealed FSGS in all. All TRIM8 variants cause protein truncation clustering within the last exon between residues 390 and 487 of the 551 amino acid protein, indicating a correlation between this syndrome and loss of the TRIM8 C-terminal region. Wild-type TRIM8 overexpressed in immortalized human podocytes and neuronal cells localized to nuclear bodies, while constructs harboring patient-specific variants mislocalized diffusely to the nucleoplasm. Co-localization studies demonstrated that Gemini and Cajal bodies frequently abut a TRIM8 nuclear body. Truncating TRIM8 DNVs cause a neuro-renal syndrome via aberrant TRIM8 localization, implicating nuclear bodies in FSGS and developmental brain disease.


Sujet(s)
Protéines de transport/génétique , Incapacités de développement/génétique , Épilepsie/génétique , Glomérulonéphrite segmentaire et focale/génétique , Espace intranucléaire/métabolisme , Syndrome néphrotique/génétique , Syndrome néphrotique/métabolisme , Protéines de tissu nerveux/génétique , Adulte , Animaux , Protéines de transport/composition chimique , Protéines de transport/métabolisme , Lignée cellulaire , Enfant , Enfant d'âge préscolaire , Codon non-sens , Incapacités de développement/métabolisme , Épilepsie/métabolisme , Femelle , Glomérulonéphrite segmentaire et focale/métabolisme , Humains , Rein/métabolisme , Mâle , Souris , Mutation , Protéines de tissu nerveux/composition chimique , Protéines de tissu nerveux/métabolisme , Phénotype , Podocytes/métabolisme ,
11.
Nephrol Dial Transplant ; 36(2): 237-246, 2021 01 25.
Article de Anglais | MEDLINE | ID: mdl-33097957

RÉSUMÉ

BACKGROUND: An underlying monogenic cause of early-onset chronic kidney disease (CKD) can be detected in ∼20% of individuals. For many etiologies of CKD manifesting before 25 years of age, >200 monogenic causative genes have been identified to date, leading to the elucidation of mechanisms of renal pathogenesis. METHODS: In 51 families with echogenic kidneys and CKD, we performed whole-exome sequencing to identify novel monogenic causes of CKD. RESULTS: We discovered a homozygous truncating mutation in the transcription factor gene transcription factor CP2-like 1 (TFCP2L1) in an Arabic patient of consanguineous descent. The patient developed CKD by the age of 2 months and had episodes of severe hypochloremic, hyponatremic and hypokalemic alkalosis, seizures, developmental delay and hypotonia together with cataracts. We found that TFCP2L1 was localized throughout kidney development particularly in the distal nephron. Interestingly, TFCP2L1 induced the growth and development of renal tubules from rat mesenchymal cells. Conversely, the deletion of TFCP2L1 in mice was previously shown to lead to reduced expression of renal cell markers including ion transporters and cell identity proteins expressed in different segments of the distal nephron. TFCP2L1 localized to the nucleus in HEK293T cells only upon coexpression with its paralog upstream-binding protein 1 (UBP1). A TFCP2L1 mutant complementary DNA (cDNA) clone that represented the patient's mutation failed to form homo- and heterodimers with UBP1, an essential step for its transcriptional activity. CONCLUSION: Here, we identified a loss-of-function TFCP2L1 mutation as a potential novel cause of CKD in childhood accompanied by a salt-losing tubulopathy.


Sujet(s)
Transition épithélio-mésenchymateuse , Maladies du rein/étiologie , Mutation , Protéines de répression/génétique , Animaux , Enfant , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Femelle , Cellules HEK293 , Humains , Maladies du rein/métabolisme , Maladies du rein/anatomopathologie , Souris , Souris knockout , Rats , Protéines de répression/métabolisme , Analyse sur cellule unique , Facteurs de transcription/génétique , Facteurs de transcription/métabolisme ,
12.
Pediatr Nephrol ; 36(2): 463-471, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-32715379

RÉSUMÉ

BACKGROUND: Atypical hemolytic uremic syndrome (aHUS) is a rare, life-threatening microangiopathy, frequently causing kidney failure. Inhibition of the terminal complement complex with eculizumab is the only licensed treatment but mostly requires long-term administration and risks severe side effects. The underlying genetic cause of aHUS is thought to influence the severity of initial and recurring episodes, with milder courses in patients with mutations in membrane cofactor protein (MCP). METHODS: Twenty pediatric cases of aHUS due to isolated heterozygous MCP mutations were reported from 12 German pediatric nephrology centers to describe initial presentation, timing of relapses, treatment, and kidney outcome. RESULTS: The median age of onset was 4.6 years, with a female to male ratio of 1:3. Without eculizumab maintenance therapy, 50% (9/18) of the patients experienced a first relapse after a median period of 3.8 years. Kaplan-Meier analysis showed a relapse-free survival of 93% at 1 year. Four patients received eculizumab long-term treatment, while 3 patients received short courses. We could not show a benefit from complement blockade therapy on long term kidney function, independent of short-term or long-term treatment. To prevent 1 relapse with eculizumab, the theoretical number-needed-to-treat (NNT) was 15 for the first year and 3 for the first 5 years after initial presentation. CONCLUSION: Our study shows that heterozygous MCP mutations cause aHUS with a risk of first relapse of about 10% per year, resulting in large NNTs for prevention of relapses with eculizumab. More studies are needed to define an optimal treatment schedule for patients with MCP mutations to minimize the risks of the disease and treatment.


Sujet(s)
Syndrome hémolytique et urémique atypique , Défaillance rénale chronique , Syndrome hémolytique et urémique atypique/traitement médicamenteux , Syndrome hémolytique et urémique atypique/génétique , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Mâle , Antigènes CD46 , Mutation , Récidive
13.
Am J Hum Genet ; 107(6): 1113-1128, 2020 12 03.
Article de Anglais | MEDLINE | ID: mdl-33232676

RÉSUMÉ

The discovery of >60 monogenic causes of nephrotic syndrome (NS) has revealed a central role for the actin regulators RhoA/Rac1/Cdc42 and their effectors, including the formin INF2. By whole-exome sequencing (WES), we here discovered bi-allelic variants in the formin DAAM2 in four unrelated families with steroid-resistant NS. We show that DAAM2 localizes to the cytoplasm in podocytes and in kidney sections. Further, the variants impair DAAM2-dependent actin remodeling processes: wild-type DAAM2 cDNA, but not cDNA representing missense variants found in individuals with NS, rescued reduced podocyte migration rate (PMR) and restored reduced filopodia formation in shRNA-induced DAAM2-knockdown podocytes. Filopodia restoration was also induced by the formin-activating molecule IMM-01. DAAM2 also co-localizes and co-immunoprecipitates with INF2, which is intriguing since variants in both formins cause NS. Using in vitro bulk and TIRF microscopy assays, we find that DAAM2 variants alter actin assembly activities of the formin. In a Xenopus daam2-CRISPR knockout model, we demonstrate actin dysregulation in vivo and glomerular maldevelopment that is rescued by WT-DAAM2 mRNA. We conclude that DAAM2 variants are a likely cause of monogenic human SRNS due to actin dysregulation in podocytes. Further, we provide evidence that DAAM2-associated SRNS may be amenable to treatment using actin regulating compounds.


Sujet(s)
Actines/métabolisme , Variation génétique , Protéines des microfilaments/génétique , Syndrome néphrotique/génétique , Protéines G rho/génétique , Allèles , Animaux , Animal génétiquement modifié , Mouvement cellulaire/génétique , Cytoplasme/métabolisme , Formines/métabolisme , Humains , Rein/métabolisme , Glomérule rénal/métabolisme , Mutation faux-sens , Podocytes/métabolisme , Pseudopodes/métabolisme , Petit ARN interférent/métabolisme , , Xenopus
14.
Am J Hum Genet ; 107(4): 727-742, 2020 10 01.
Article de Anglais | MEDLINE | ID: mdl-32891193

RÉSUMÉ

Congenital anomalies of the kidney and urinary tract (CAKUT) constitute one of the most frequent birth defects and represent the most common cause of chronic kidney disease in the first three decades of life. Despite the discovery of dozens of monogenic causes of CAKUT, most pathogenic pathways remain elusive. We performed whole-exome sequencing (WES) in 551 individuals with CAKUT and identified a heterozygous de novo stop-gain variant in ZMYM2 in two different families with CAKUT. Through collaboration, we identified in total 14 different heterozygous loss-of-function mutations in ZMYM2 in 15 unrelated families. Most mutations occurred de novo, indicating possible interference with reproductive function. Human disease features are replicated in X. tropicalis larvae with morpholino knockdowns, in which expression of truncated ZMYM2 proteins, based on individual mutations, failed to rescue renal and craniofacial defects. Moreover, heterozygous Zmym2-deficient mice recapitulated features of CAKUT with high penetrance. The ZMYM2 protein is a component of a transcriptional corepressor complex recently linked to the silencing of developmentally regulated endogenous retrovirus elements. Using protein-protein interaction assays, we show that ZMYM2 interacts with additional epigenetic silencing complexes, as well as confirming that it binds to FOXP1, a transcription factor that has also been linked to CAKUT. In summary, our findings establish that loss-of-function mutations of ZMYM2, and potentially that of other proteins in its interactome, as causes of human CAKUT, offering new routes for studying the pathogenesis of the disorder.


Sujet(s)
Protéines de liaison à l'ADN/génétique , Épigenèse génétique , Facteurs de transcription Forkhead/génétique , Mutation , Protéines de répression/génétique , Facteurs de transcription/génétique , Voies urinaires/métabolisme , Malformations urogénitales/génétique , Protéines d'amphibien/antagonistes et inhibiteurs , Protéines d'amphibien/génétique , Protéines d'amphibien/métabolisme , Animaux , Études cas-témoins , Enfant , Enfant d'âge préscolaire , Protéines de liaison à l'ADN/métabolisme , Famille , Femelle , Facteurs de transcription Forkhead/métabolisme , Hétérozygote , Humains , Nourrisson , Larve/génétique , Larve/croissance et développement , Larve/métabolisme , Mâle , Souris , Souris knockout , Morpholinos/génétique , Morpholinos/métabolisme , Pedigree , Liaison aux protéines , Protéines de répression/métabolisme , Facteurs de transcription/métabolisme , Voies urinaires/malformations , Malformations urogénitales/métabolisme , Malformations urogénitales/anatomopathologie , , Xenopus
15.
Genet Med ; 22(10): 1673-1681, 2020 10.
Article de Anglais | MEDLINE | ID: mdl-32475988

RÉSUMÉ

PURPOSE: Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney disease in childhood and adolescence. We aim to identify novel monogenic causes of CAKUT. METHODS: Exome sequencing was performed in 550 CAKUT-affected families. RESULTS: We discovered seven FOXC1 heterozygous likely pathogenic variants within eight CAKUT families. These variants are either never reported, or present in <5 alleles in the gnomAD database with ~141,456 controls. FOXC1 is a causal gene for Axenfeld-Rieger syndrome type 3 and anterior segment dysgenesis 3. Pathogenic variants in FOXC1 have not been detected in patients with CAKUT yet. Interestingly, mouse models for Foxc1 show severe CAKUT phenotypes with incomplete penetrance and variable expressivity. The FOXC1 variants are enriched in the CAKUT cohort compared with the control. Genotype-phenotype correlations showed that Axenfeld-Rieger syndrome or anterior segment dysgenesis can be caused by both truncating and missense pathogenic variants, and the missense variants are located at the forkhead domain. In contrast, for CAKUT, there is no truncating pathogenic variant, and all variants except one are located outside the forkhead domain. CONCLUSION: We thereby expanded the phenotype of FOXC1 pathogenic variants toward involvement of CAKUT, which can potentially be explained by allelism.


Sujet(s)
Malformations oculaires , Voies urinaires , Enfant , Facteurs de transcription Forkhead/génétique , Hétérozygote , Humains , Rein , Phénotype
16.
Nat Biomed Eng ; 4(6): 601-609, 2020 06.
Article de Anglais | MEDLINE | ID: mdl-32284553

RÉSUMÉ

In organ transplantation, infection and rejection are major causes of graft loss. They are linked by the net state of immunosuppression. To diagnose and treat these conditions earlier, and to improve long-term patient outcomes, refined strategies for the monitoring of patients after graft transplantation are needed. Here, we show that a fast and inexpensive assay based on CRISPR-Cas13 accurately detects BK polyomavirus DNA and cytomegalovirus DNA from patient-derived blood and urine samples, as well as CXCL9 messenger RNA (a marker of graft rejection) at elevated levels in urine samples from patients experiencing acute kidney transplant rejection. The assay, which we adapted for lateral-flow readout, enables-via simple visualization-the post-transplantation monitoring of common opportunistic viral infections and of graft rejection, and should facilitate point-of-care post-transplantation monitoring.


Sujet(s)
Systèmes CRISPR-Cas , Rejet du greffon/virologie , Infections opportunistes/diagnostic , Anatomopathologie moléculaire/méthodes , Marqueurs biologiques/sang , Marqueurs biologiques/urine , Chimiokine CXCL9/sang , Chimiokine CXCL9/urine , Clustered regularly interspaced short palindromic repeats , Cytomegalovirus/génétique , Cytomegalovirus/isolement et purification , Infections à cytomégalovirus/diagnostic , ADN viral/sang , ADN viral/génétique , ADN viral/urine , Humains , Rein , Maladies du rein/virologie , Transplantation rénale/effets indésirables , Mâle , Adulte d'âge moyen , Analyse sur le lieu d'intervention , Polyomavirus/génétique , Polyomavirus/isolement et purification , Infections à polyomavirus/diagnostic , Complications postopératoires/diagnostic , ARN messager , Infections à virus oncogènes/diagnostic
17.
Kidney Int ; 97(3): 567-579, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31959358

RÉSUMÉ

Distal renal tubular acidosis is a rare renal tubular disorder characterized by hyperchloremic metabolic acidosis and impaired urinary acidification. Mutations in three genes (ATP6V0A4, ATP6V1B1 and SLC4A1) constitute a monogenic causation in 58-70% of familial cases of distal renal tubular acidosis. Recently, mutations in FOXI1 have been identified as an additional cause. Therefore, we hypothesized that further monogenic causes of distal renal tubular acidosis remain to be discovered. Panel sequencing and/or whole exome sequencing was performed in a cohort of 17 families with 19 affected individuals with pediatric onset distal renal tubular acidosis. A causative mutation was detected in one of the three "classical" known distal renal tubular acidosis genes in 10 of 17 families. The seven unsolved families were then subjected to candidate whole exome sequencing analysis. Potential disease causing mutations in three genes were detected: ATP6V1C2, which encodes another kidney specific subunit of the V-type proton ATPase (1 family); WDR72 (2 families), previously implicated in V-ATPase trafficking in cells; and SLC4A2 (1 family), a paralog of the known distal renal tubular acidosis gene SLC4A1. Two of these mutations were assessed for deleteriousness through functional studies. Yeast growth assays for ATP6V1C2 revealed loss-of-function for the patient mutation, strongly supporting ATP6V1C2 as a novel distal renal tubular acidosis gene. Thus, we provided a molecular diagnosis in a known distal renal tubular acidosis gene in 10 of 17 families (59%) with this disease, identified mutations in ATP6V1C2 as a novel human candidate gene, and provided further evidence for phenotypic expansion in WDR72 mutations from amelogenesis imperfecta to distal renal tubular acidosis.


Sujet(s)
Acidose tubulaire rénale , Vacuolar Proton-Translocating ATPases , Acidose tubulaire rénale/génétique , Protéine érythrocytaire-1 échangeuse d'anions , Enfant , Antiporteurs des ions chlorure-bicarbonate , Analyse de mutations d'ADN , Facteurs de transcription Forkhead , Humains , Mutation , Vacuolar Proton-Translocating ATPases/génétique ,
18.
Am J Hum Genet ; 105(6): 1286-1293, 2019 12 05.
Article de Anglais | MEDLINE | ID: mdl-31708116

RÉSUMÉ

Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney disease in the first three decades of life, and in utero obstruction to urine flow is a frequent cause of secondary upper urinary tract malformations. Here, using whole-exome sequencing, we identified three different biallelic mutations in CHRNA3, which encodes the α3 subunit of the nicotinic acetylcholine receptor, in five affected individuals from three unrelated families with functional lower urinary tract obstruction and secondary CAKUT. Four individuals from two families have additional dysautonomic features, including impaired pupillary light reflexes. Functional studies in vitro demonstrated that the mutant nicotinic acetylcholine receptors were unable to generate current following stimulation with acetylcholine. Moreover, the truncating mutations p.Thr337Asnfs∗81 and p.Ser340∗ led to impaired plasma membrane localization of CHRNA3. Although the importance of acetylcholine signaling in normal bladder function has been recognized, we demonstrate for the first time that mutations in CHRNA3 can cause bladder dysfunction, urinary tract malformations, and dysautonomia. These data point to a pathophysiologic sequence by which monogenic mutations in genes that regulate bladder innervation may secondarily cause CAKUT.


Sujet(s)
Maladies du système nerveux autonome/étiologie , Rein/malformations , Mutation , Récepteurs nicotiniques/génétique , Voies urinaires/malformations , Malformations urogénitales/étiologie , Adulte , Maladies du système nerveux autonome/génétique , Maladies du système nerveux autonome/anatomopathologie , Femelle , Études de suivi , Humains , Rein/anatomopathologie , Mâle , Pedigree , Pronostic , Voies urinaires/anatomopathologie , Malformations urogénitales/génétique , Malformations urogénitales/anatomopathologie , Jeune adulte
19.
Pediatr Transplant ; 22(4): e13195, 2018 06.
Article de Anglais | MEDLINE | ID: mdl-29665156

RÉSUMÉ

HEV infection can lead to chronic hepatitis in immunosuppressed patients; extrahepatic manifestations are rarely seen. Here, we report a 13-year-old renal transplant patient with chronic hepatitis E and renal involvement. Ribavirin therapy led to temporary virus clearance and amelioration of kidney function. However, ribavirin therapy caused severe hyporegenerative anemia, which has so far only been reported in patients treated with a combination of ribavirin and interferon alpha.


Sujet(s)
Anémie/induit chimiquement , Antiviraux/effets indésirables , Hépatite E/traitement médicamenteux , Transplantation rénale , Complications postopératoires/traitement médicamenteux , Ribavirine/effets indésirables , Adolescent , Anémie/diagnostic , Antiviraux/usage thérapeutique , Maladie chronique , Femelle , Hépatite E/étiologie , Humains , Ribavirine/usage thérapeutique
20.
J Immunol ; 195(12): 5795-804, 2015 Dec 15.
Article de Anglais | MEDLINE | ID: mdl-26538394

RÉSUMÉ

Acute graft-versus-host disease (GvHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation. During the initiation phase of acute GvHD, endogenous danger signals such as ATP are released and inform the innate immune system via activation of the purinergic receptor P2X7 that a noninfectious damage has occurred. A second ATP-activated purinergic receptor involved in inflammatory diseases is P2Y2. In this study, we used P2y2(-/-) mice to test the role of this receptor in GvHD. P2y2(-/-) recipients experienced reduced GvHD-related mortality, IL-6 levels, enterocyte apoptosis, and histopathology scores. Chimeric mice with P2y2 deficiency restricted to hematopoietic tissues survived longer after GvHD induction than did wild-type mice. P2y2 deficiency of the recipient was connected to lower levels of myeloperoxidase in the intestinal tract of mice developing GvHD and a reduced myeloid cell signature. Selective deficiency of P2Y2 in inflammatory monocytes decreased GvHD severity. Mechanistically, P2y2(-/-) inflammatory monocytes displayed defective ERK activation and reactive oxygen species production. Compatible with a role of P2Y2 in human GvHD, the frequency of P2Y2(+) cells in inflamed GvHD lesions correlated with histopathological GvHD severity. Our findings indicate a novel function for P2Y2 in ATP-activated recipient myeloid cells during GvHD, which could be exploited when targeting danger signals to prevent GvHD.


Sujet(s)
Maladie du greffon contre l'hôte/immunologie , Transplantation de cellules souches hématopoïétiques , Cellules myéloïdes/métabolisme , Récepteurs purinergiques P2Y2/métabolisme , Adénosine triphosphate/métabolisme , Animaux , Maladie du greffon contre l'hôte/traitement médicamenteux , Humains , Interleukine-6/métabolisme , Intestins/immunologie , Système de signalisation des MAP kinases/génétique , Souris , Souris de lignée C57BL , Souris knockout , Thérapie moléculaire ciblée , Cellules myéloïdes/immunologie , Espèces réactives de l'oxygène/métabolisme , Récepteurs purinergiques P2Y2/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE