Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Cells ; 13(2)2024 01 12.
Article in English | MEDLINE | ID: mdl-38247840

ABSTRACT

Besides visceral heterotaxia, Pkd1l1 null mouse embryos exhibit general edema and perinatal lethality. In humans, congenital chylothorax (CCT) is a frequent cause of fetal hydrops. In 2021, Correa and colleagues reported ultrarare compound heterozygous variants in PKD1L1 exhibiting in two consecutive fetuses with severe hydrops, implicating a direct role of PKD1L1 in fetal hydrops formation. Here, we performed an exome survey and identified ultrarare compound heterozygous variants in PKD1L1 in two of the five case-parent trios with CCT. In one family, the affected carried the ultrarare missense variants c.1543G>A(p.Gly515Arg) and c.3845T>A(p.Val1282Glu). In the other family, the affected carried the ultrarare loss-of-function variant (LoF) c.863delA(p.Asn288Thrfs*3) and the ultrarare missense variant c.6549G>T(p.Gln2183His). Investigation of the variants' impact on PKD1L1 protein localization suggests the missense variants cause protein dysfunction and the LoF variant causes protein mislocalization. Further analysis of Pkd1l1 mutant mouse embryos revealed about 20% of Pkd1l1-/- embryos display general edema and pleural effusion at 14.5 dpc. Immunofluorescence staining at 14.5 dpc in Pkd1l1-/- embryos displayed both normal and massively altered lymphatic vessel morphologies. Together, our studies suggest the implication of PKD1L1 in congenital lymphatic anomalies, including CCTs.


Subject(s)
Chylothorax , Animals , Female , Humans , Mice , Pregnancy , Chylothorax/genetics , Fetus , Genetic Diseases, X-Linked , Hydrops Fetalis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Knockout
2.
J Med Genet ; 60(6): 587-596, 2023 06.
Article in English | MEDLINE | ID: mdl-36379543

ABSTRACT

BACKGROUND: SHROOM4 is thought to play an important role in cytoskeletal modification and development of the early nervous system. Previously, single-nucleotide variants (SNVs) or copy number variations (CNVs) in SHROOM4 have been associated with the neurodevelopmental disorder Stocco dos Santos syndrome, but not with congenital anomalies of the urinary tract and the visceral or the cardiovascular system. METHODS: Here, exome sequencing and CNV analyses besides expression studies in zebrafish and mouse and knockdown (KD) experiments using a splice blocking morpholino in zebrafish were performed to study the role of SHROOM4 during embryonic development. RESULTS: In this study, we identified putative disease-causing SNVs and CNVs in SHROOM4 in six individuals from four families with congenital anomalies of the urinary tract and the anorectal, cardiovascular and central nervous systems (CNS). Embryonic mouse and zebrafish expression studies showed Shroom4 expression in the upper and lower urinary tract, the developing cloaca, the heart and the cerebral CNS. KD studies in zebrafish larvae revealed pronephric cysts, anomalies of the cloaca and the heart, decreased eye-to-head ratio and higher mortality compared with controls. These phenotypes could be rescued by co-injection of human wild-type SHROOM4 mRNA and morpholino. CONCLUSION: The identified SNVs and CNVs in affected individuals with congenital anomalies of the urinary tract, the anorectal, the cardiovascular and the central nervous systems, and subsequent embryonic mouse and zebrafish studies suggest SHROOM4 as a developmental gene for different organ systems.


Subject(s)
Cardiovascular System , Urinary Tract , Pregnancy , Female , Humans , Animals , Mice , Zebrafish/genetics , DNA Copy Number Variations , Morpholinos , Urinary Tract/abnormalities , Central Nervous System
4.
Genes (Basel) ; 12(9)2021 08 24.
Article in English | MEDLINE | ID: mdl-34573284

ABSTRACT

Anorectal malformations (ARM) represent a rare birth defect of the hindgut that occur in approximately 1 in 3000 live births. Around 60% of ARM occur with associated anomalies including defined genetic syndromes and associations with chromosomal aberrations. The etiology of ARM is heterogeneous, with the individual environmental or genetic risk factors remaining unknown for the majority of cases. The occurrence of familial ARM and previous epidemiologic analysis suggest autosomal dominant inheritance in a substantial subset of ARM patients. The implicated mortality and reduced fecundity in patients with ARM would lead to allele loss. However, mutational de novo events among the affected individuals could compensate for the evolutionary pressure. With the implementation of exome sequencing, array-based molecular karyotyping and family-based rare variant analyses, the technologies are available to identify the respective factors. This review discusses the identification of disease-causing variants among individuals with ARM. It highlights the role of mutational de novo events.


Subject(s)
Anorectal Malformations/genetics , DNA Copy Number Variations , Anorectal Malformations/diagnosis , Anorectal Malformations/epidemiology , Genetic Loci , Genetic Testing/methods , Humans , Mutation
6.
Orphanet J Rare Dis ; 16(1): 167, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33836786

ABSTRACT

BACKGROUND: The triad of a presacral mass, sacral agenesis and an anorectal anomaly constitutes the rare Currarino syndrome (CS), which is caused by dorsal-ventral patterning defects during embryonic development. The major causative CS gene is MNX1, encoding a homeobox protein. MAIN BODY: In the majority of patients, CS occurs as an autosomal dominant trait; however, a female predominance observed, implies that CS may underlie an additional mode(s) of inheritance. Often, the diagnosis of CS is established solely by clinical findings, impacting a detailed analysis of the disease. Our combined data, evaluating more than 60 studies reporting patients with CS-associated mutations, revealed a slightly higher incidence rate in females with a female-to-male ratio of 1.39:1. Overall, MNX1 mutation analysis was successful in only 57.4% of all CS patients investigated, with no mutation detected in 7.7% of the familial and 68% of the sporadic patients. Our studies failed to detect the presence of an expressed MNX1 isoform that might explain at least some of these mutation-negative cases. CONCLUSION: Aside from MNX1, other genes or regulatory regions may contribute to CS and we discuss several cytogenetic studies and whole-exome sequencing data that have implicated further loci/genes in its etiology.


Subject(s)
Digestive System Abnormalities , Syringomyelia , Anal Canal/abnormalities , Digestive System Abnormalities/genetics , Female , Homeodomain Proteins/genetics , Humans , Male , Rectum/abnormalities , Sacrum/abnormalities , Transcription Factors/genetics
7.
Front Pediatr ; 8: 310, 2020.
Article in English | MEDLINE | ID: mdl-32656166

ABSTRACT

Background: The VATER/VACTERL association (VACTERL) is defined as the non-random occurrence of the following congenital anomalies: Vertebral, Anal, Cardiac, Tracheal-Esophageal, Renal, and Limb anomalies. As no unequivocal candidate gene has been identified yet, patients are diagnosed phenotypically. The aims of this study were to identify patients with monogenic disorders using a genetics-first approach, and to study whether variants in candidate genes are involved in the etiology of VACTERL or the individual features of VACTERL: Anorectal malformation (ARM) or esophageal atresia with or without trachea-esophageal fistula (EA/TEF). Methods: Using molecular inversion probes, a candidate gene panel of 56 genes was sequenced in three patient groups: VACTERL (n = 211), ARM (n = 204), and EA/TEF (n = 95). Loss-of-function (LoF) and additional likely pathogenic missense variants, were prioritized and validated using Sanger sequencing. Validated variants were tested for segregation and patients were clinically re-evaluated. Results: In 7 out of the 510 patients (1.4%), pathogenic or likely pathogenic variants were identified in SALL1, SALL4, and MID1, genes that are associated with Townes-Brocks, Duane-radial-ray, and Opitz-G/BBB syndrome. These syndromes always include ARM or EA/TEF, in combination with at least two other VACTERL features. We did not identify LoF variants in the remaining candidate genes. Conclusions: None of the other candidate genes were identified as novel unequivocal disease genes for VACTERL. However, a genetics-first approach allowed refinement of the clinical diagnosis in seven patients, in whom an alternative molecular-based diagnosis was found with important implications for the counseling of the families.

8.
Front Pediatr ; 8: 174, 2020.
Article in English | MEDLINE | ID: mdl-32509709

ABSTRACT

Introduction: To evaluate the impact of reconstructive strategies and post-operative management on short- and long-term surgical outcome and complications of classical bladder exstrophy (CBE) patients' comprehensive data of the multicenter German-wide Network for Congenital Uro-Rectal malformations (CURE-Net) were analyzed. Methods: Descriptive analyses were performed between 34 prospectively collected CBE patients born since 2009, median 3 months old [interquartile range (IQR), 2-4 months], and 113 cross-sectional patients, median 12 years old (IQR, 6-21 years). Results: The majority of included individuals were males (67%). Sixty-eight percent of the prospectively observed and 53% of the cross-sectional patients were reconstructed using a staged approach (p = 0.17). Although prospectively observed patients were operated on at a younger age, the post-operative management did not significantly change in the years before and after 2009. Solely, in prospectively observed patients, peridural catheters were used significantly more often (p = 0.017). Blood transfusions were significantly more frequent in males (p = 0.002). Only half of all CBE individuals underwent inguinal hernia repair. Cross-sectional patients after single-stage reconstructions showed more direct post-operative complications such as upper urinary tract dilatations (p = 0.0021) or urinary tract infections (p = 0.023), but not more frequent renal function impairment compared to patients after the staged approach (p = 0.42). Continence outcomes were not significantly different between the concepts (p = 0.51). Self-reported continence data showed that the majority of the included CBE patients was intermittent or continuous incontinent. Furthermore, subsequent consecutive augmentations and catheterizable stomata did not significantly differ between the two operative approaches. Urinary diversions were only reported after the staged concept. Conclusions: In this German multicenter study, a trend toward the staged concept was observed. While single-stage approaches tended to have initially more complications such as renal dilatation or urinary tract infections, additional surgery such as augmentations and stomata appeared to be similar after staged and single-stage reconstructions in the long term.

9.
Sci Rep ; 8(1): 10735, 2018 Jul 16.
Article in English | MEDLINE | ID: mdl-30013141

ABSTRACT

Pulmonary hypertension (PH) contributes to high mortality in congenital diaphragmatic hernia (CDH). A better understanding of the regulatory mechanisms underlying the pathology in CDH might allow the identification of prognostic biomarkers and potential therapeutic targets. We report the results from an expression profiling of circulating microRNAs (miRNAs) in direct post-pulmonary blood flow of 18 CDH newborns. Seven miRNAs differentially expressed in children that either died or developed chronic lung disease (CLD) up to 28 days after birth, compared to those who survived without developing CLD during this period, were identified. Target gene and pathway analyses indicate that these miRNAs functions include regulation of the cell cycle, inflammation and morphogenesis, by targeting molecules responsive to growth factors, cytokines and cellular stressors. Furthermore, we identified hub molecules by constructing a protein-protein interaction network of shared targets, and ranked the relative importance of the identified miRNAs. Our results suggest that dysregulations in miRNAs let-7b-5p, -7c-5p, miR-1307-3p, -185-3p, -8084, -331-3p and -210-3p may be detrimental for the development and function of the lungs and pulmonary vasculature, compromise cardiac function and contribute to the development of CLD in CDH. Further investigation of the biomarker and therapeutic potential of these circulating miRNAs is encouraged.


Subject(s)
Circulating MicroRNA/metabolism , Gene Regulatory Networks , Hernias, Diaphragmatic, Congenital/complications , Hypertension, Pulmonary/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Biomarkers/blood , Circulating MicroRNA/blood , Female , Gene Expression Profiling , Gene Expression Regulation , Hernias, Diaphragmatic, Congenital/blood , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/diagnosis , Infant, Newborn , Male , Prognosis , Prospective Studies , Protein Interaction Mapping , Protein Interaction Maps , Pulmonary Disease, Chronic Obstructive/blood , Pulmonary Disease, Chronic Obstructive/diagnosis
10.
Cell Metab ; 24(2): 234-45, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27508872

ABSTRACT

Human genetics studies have implicated GALNT2, encoding GalNAc-T2, as a regulator of high-density lipoprotein cholesterol (HDL-C) metabolism, but the mechanisms relating GALNT2 to HDL-C remain unclear. We investigated the impact of homozygous GALNT2 deficiency on HDL-C in humans and mammalian models. We identified two humans homozygous for loss-of-function mutations in GALNT2 who demonstrated low HDL-C. We also found that GALNT2 loss of function in mice, rats, and nonhuman primates decreased HDL-C. O-glycoproteomics studies of a human GALNT2-deficient subject validated ANGPTL3 and ApoC-III as GalNAc-T2 targets. Additional glycoproteomics in rodents identified targets influencing HDL-C, including phospholipid transfer protein (PLTP). GALNT2 deficiency reduced plasma PLTP activity in humans and rodents, and in mice this was rescued by reconstitution of hepatic Galnt2. We also found that GALNT2 GWAS SNPs associated with reduced HDL-C also correlate with lower hepatic GALNT2 expression. These results posit GALNT2 as a direct modulator of HDL metabolism across mammals.


Subject(s)
Lipoproteins, HDL/metabolism , N-Acetylgalactosaminyltransferases/deficiency , Amino Acid Sequence , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/metabolism , Animals , Base Sequence , Cholesterol, HDL/blood , Gene Knockdown Techniques , Glycoproteins/metabolism , Homozygote , Humans , Liver/enzymology , Mice , Mice, Knockout , Models, Animal , Mutation/genetics , N-Acetylgalactosaminyltransferases/chemistry , N-Acetylgalactosaminyltransferases/genetics , N-Acetylgalactosaminyltransferases/metabolism , Phenotype , Phospholipid Transfer Proteins/metabolism , Polymorphism, Single Nucleotide/genetics , Primates , Proteomics , Rats , Triglycerides/metabolism , Polypeptide N-acetylgalactosaminyltransferase
11.
Eur J Pediatr ; 175(6): 825-31, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26979529

ABSTRACT

UNLABELLED: The purpose of our study was to investigate the importance of amniotic fluid (AF) for fetal growth during late gestation using esophageal atresia (EA) patients as a model. In this retrospective cohort study, we compared the z-scores adapted for birth weights (BW z-scores) for each of 517 European newborns with congenital pre-gastric intestinal atresia, i.e., EA, to a European reference population. To account for the influence of the intestinal atresia on fetal growth per se, we compared adapted birth weights for each of 504 European newborns with post colonic intestinal atresia (anorectal malformation (ARM) with atresia of the anus) to the same European reference population. Analysis of the complete cohort showed (i) a significantly higher rate of small for gestational age newborns among EA compared to ARM newborns (p < 0.001) and (ii) significantly lower BW z-scores among EA compared to ARM newborns (p < 0.001). BW z-scores of EA newborns were significantly lower in term compared to preterm newborns with an inverse correlation with gestational age (GA) (Spearman correlation coefficient, r = -0.185, p < 0.001). CONCLUSIONS: Enteral uptake of AF seems to play a pivotal role in fetal growth during late gestation. WHAT IS KNOWN: • Peak velocity of fetal weight gain occurs at 33 weeks of gestation and continues until birth. During this period, fetal growth is mainly characterized by cellular hypertrophy. • Amniotic fluid (AF) comprises large amounts of hormones and growth regulators. What is New: • A significantly higher rate of small for gestational age and lower birth weights and z-scores are observed among newborn infants with congenital pre-gastric intestinal atresia. • These findings suggest that enteral uptake of AF is a major predictor for fetal growth during late gestation.


Subject(s)
Amniotic Fluid/physiology , Birth Weight/physiology , Colon/abnormalities , Esophageal Atresia/physiopathology , Fetal Development , Infant, Small for Gestational Age/physiology , Intestinal Atresia/physiopathology , Anorectal Malformations/physiopathology , Case-Control Studies , Female , Gestational Age , Humans , Infant, Newborn , Male , Pregnancy , Retrospective Studies , Sex Distribution , Statistics, Nonparametric
12.
Nephrol Dial Transplant ; 31(8): 1280-3, 2016 08.
Article in English | MEDLINE | ID: mdl-26908769

ABSTRACT

BACKGROUND: Congenital anomalies of the kidney and urinary tract (CAKUT) are the most common cause of chronic kidney diseases in children and young adults, accounting for ∼50% of cases. These anomalies represent maldevelopment of the genitourinary system and can be genetically explained in only 10-16% of cases by mutations or by copy number variations in protein coding sequences. Knock-out mouse models, lacking components of the microRNA (miRNA) processing machinery (i.e. Dicer, Drosha, Dgcr8), exhibit kidney malformations resembling human CAKUT. METHODS: Given the Dicer-null mouse phenotype, which implicates a central role for miRNAs gene regulation during kidney development, we hypothesized that miRNAs expressed during kidney development may cause CAKUT in humans if mutated. To evaluate this possibility we carried out Next-Generation sequencing of 96 stem-loop regions of 73 renal developmental miRNA genes in 1248 individuals with non-syndromic CAKUT from 980 families. RESULTS: We sequenced 96 stem-loop regions encoded by 73 miRNA genes that are expressed during kidney development in humans, mice and rats. Overall, we identified in 31/1213 individuals from 26 families with 17 different single nucleotide variants. Two variants did not segregate with the disease and hence were not causative. Thirteen variants were likely benign variants because they occurred in control populations and/or they affected nucleotides of weak evolutionary conservation. Two out of 1213 unrelated individuals had potentially pathogenic variants with unknown biologic relevance affecting miRNAs MIR19B1 and MIR99A. CONCLUSIONS: Our results indicate that mutations affecting mature microRNAs in individuals with CAKUT are rare and thus most likely not a common cause of CAKUT in humans.


Subject(s)
Kidney/abnormalities , MicroRNAs/genetics , Mutation , Urinary Tract/abnormalities , Urogenital Abnormalities/genetics , Adolescent , Animals , Child , DNA Copy Number Variations , Humans , Mice , Mice, Knockout , Phenotype , Rats , Young Adult
13.
Am J Hum Genet ; 97(2): 291-301, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26235987

ABSTRACT

Congenital anomalies of the kidneys and urinary tract (CAKUT) are the most common cause of chronic kidney disease in the first three decades of life. Identification of single-gene mutations that cause CAKUT permits the first insights into related disease mechanisms. However, for most cases the underlying defect remains elusive. We identified a kindred with an autosomal-dominant form of CAKUT with predominant ureteropelvic junction obstruction. By whole exome sequencing, we identified a heterozygous truncating mutation (c.1010delG) of T-Box transcription factor 18 (TBX18) in seven affected members of the large kindred. A screen of additional families with CAKUT identified three families harboring two heterozygous TBX18 mutations (c.1570C>T and c.487A>G). TBX18 is essential for developmental specification of the ureteric mesenchyme and ureteric smooth muscle cells. We found that all three TBX18 altered proteins still dimerized with the wild-type protein but had prolonged protein half life and exhibited reduced transcriptional repression activity compared to wild-type TBX18. The p.Lys163Glu substitution altered an amino acid residue critical for TBX18-DNA interaction, resulting in impaired TBX18-DNA binding. These data indicate that dominant-negative TBX18 mutations cause human CAKUT by interference with TBX18 transcriptional repression, thus implicating ureter smooth muscle cell development in the pathogenesis of human CAKUT.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Genes, Dominant/genetics , Muscle, Smooth/embryology , Mutation/genetics , T-Box Domain Proteins/genetics , Ureter/embryology , Urinary Tract/abnormalities , Base Sequence , Electrophoretic Mobility Shift Assay , Exome/genetics , HEK293 Cells , Humans , Immunohistochemistry , Immunoprecipitation , Microscopy, Fluorescence , Molecular Sequence Data , Pedigree , Sequence Analysis, DNA
14.
Hum Genet ; 134(8): 905-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26026792

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) account for 40-50% of chronic kidney disease that manifests in the first two decades of life. Thus far, 31 monogenic causes of isolated CAKUT have been described, explaining ~12% of cases. To identify additional CAKUT-causing genes, we performed whole-exome sequencing followed by a genetic burden analysis in 26 genetically unsolved families with CAKUT. We identified two heterozygous mutations in SRGAP1 in 2 unrelated families. SRGAP1 is a small GTPase-activating protein in the SLIT2-ROBO2 signaling pathway, which is essential for development of the metanephric kidney. We then examined the pathway-derived candidate gene SLIT2 for mutations in cohort of 749 individuals with CAKUT and we identified 3 unrelated individuals with heterozygous mutations. The clinical phenotypes of individuals with mutations in SLIT2 or SRGAP1 were cystic dysplastic kidneys, unilateral renal agenesis, and duplicated collecting system. We show that SRGAP1 is expressed in early mouse nephrogenic mesenchyme and that it is coexpressed with ROBO2 in SIX2-positive nephron progenitor cells of the cap mesenchyme in developing rat kidney. We demonstrate that the newly identified mutations in SRGAP1 lead to an augmented inhibition of RAC1 in cultured human embryonic kidney cells and that the SLIT2 mutations compromise the ability of the SLIT2 ligand to inhibit cell migration. Thus, we report on two novel candidate genes for causing monogenic isolated CAKUT in humans.


Subject(s)
GTPase-Activating Proteins , Intercellular Signaling Peptides and Proteins , Mutation , Nerve Tissue Proteins , Receptors, Immunologic , Signal Transduction/genetics , Urogenital Abnormalities , Vesico-Ureteral Reflux , Animals , Exome , GTPase-Activating Proteins/biosynthesis , GTPase-Activating Proteins/genetics , HEK293 Cells , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Intercellular Signaling Peptides and Proteins/metabolism , Mesoderm/metabolism , Mice , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/metabolism , Rats , Receptors, Immunologic/biosynthesis , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Risk Factors , Urogenital Abnormalities/embryology , Urogenital Abnormalities/genetics , Vesico-Ureteral Reflux/embryology , Vesico-Ureteral Reflux/genetics
15.
Cardiol Young ; 25(3): 606-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24905790

ABSTRACT

VACTERL association and Scimitar syndrome are rare congenital diseases. In this study, we report on a neonate with prenatal suspicion of VACTERL association and small left-sided cardiac structures, which, only on postnatal angiography, could be revealed to be part of a Scimitar syndrome. As this is the second reported case of VACTERL association and Scimitar syndrome, the presence of Scimitar syndrome should be considered in the prenatal and postnatal evaluation of VACTERL association.


Subject(s)
Anal Canal/abnormalities , Echocardiography , Esophagus/abnormalities , Heart Defects, Congenital/diagnosis , Kidney/abnormalities , Limb Deformities, Congenital/diagnosis , Magnetic Resonance Angiography , Scimitar Syndrome/diagnosis , Spine/abnormalities , Trachea/abnormalities , Anal Canal/diagnostic imaging , Anal Canal/pathology , Comorbidity , Diagnosis, Differential , Esophagus/diagnostic imaging , Esophagus/pathology , Heart Defects, Congenital/diagnostic imaging , Heart Defects, Congenital/pathology , Humans , Infant, Newborn , Kidney/diagnostic imaging , Kidney/pathology , Limb Deformities, Congenital/diagnostic imaging , Limb Deformities, Congenital/pathology , Male , Radiography , Scimitar Syndrome/diagnostic imaging , Scimitar Syndrome/pathology , Spine/diagnostic imaging , Spine/pathology , Trachea/diagnostic imaging , Trachea/pathology , X-Rays
16.
J Am Soc Nephrol ; 25(9): 1917-22, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24700879

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) account for approximately 40% of children with ESRD in the United States. Hitherto, mutations in 23 genes have been described as causing autosomal dominant isolated CAKUT in humans. However, >90% of cases of isolated CAKUT still remain without a molecular diagnosis. Here, we hypothesized that genes mutated in recessive mouse models with the specific CAKUT phenotype of unilateral renal agenesis may also be mutated in humans with isolated CAKUT. We applied next-generation sequencing technology for targeted exon sequencing of 12 recessive murine candidate genes in 574 individuals with isolated CAKUT from 590 families. In 15 of 590 families, we identified recessive mutations in the genes FRAS1, FREM2, GRIP1, FREM1, ITGA8, and GREM1, all of which function in the interaction of the ureteric bud and the metanephric mesenchyme. We show that isolated CAKUT may be caused partially by mutations in recessive genes. Our results also indicate that biallelic missense mutations in the Fraser/MOTA/BNAR spectrum genes cause isolated CAKUT, whereas truncating mutations are found in the multiorgan form of Fraser syndrome. The newly identified recessive biallelic mutations in these six genes represent the molecular cause of isolated CAKUT in 2.5% of the 590 affected families in this study.


Subject(s)
Carrier Proteins/genetics , Extracellular Matrix Proteins/genetics , Fraser Syndrome/genetics , Integrin alpha Chains/genetics , Intercellular Signaling Peptides and Proteins/genetics , Kidney/abnormalities , Mutation , Nerve Tissue Proteins/genetics , Receptors, Interleukin/genetics , Urinary Tract/abnormalities , Vesico-Ureteral Reflux/genetics , Animals , Congenital Abnormalities/genetics , Disease Models, Animal , Female , Genes, Recessive , Humans , Kidney Diseases/congenital , Kidney Diseases/genetics , Male , Mice , Mice, Mutant Strains , Urogenital Abnormalities
17.
Kidney Int ; 85(6): 1429-33, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24429398

ABSTRACT

Congenital anomalies of the kidney and urinary tract (CAKUT) account for approximately half of children with chronic kidney disease. CAKUT can be caused by monogenic mutations; however, data are lacking on their frequency. Genetic diagnosis has been hampered by genetic heterogeneity and lack of genotype-phenotype correlation. To determine the percentage of cases with CAKUT that can be explained by mutations in known CAKUT genes, we analyzed the coding exons of the 17 known dominant CAKUT-causing genes in a cohort of 749 individuals from 650 families with CAKUT. The most common phenotypes in this CAKUT cohort were vesicoureteral reflux in 288 patients, renal hypodysplasia in 120 patients, and unilateral renal agenesis in 90 patients. We identified 37 different heterozygous mutations (33 novel) in 12 of the 17 known genes in 47 patients from 41 of the 650 families (6.3%). These mutations include (number of families): BMP7 (1), CDC5L (1), CHD1L (5), EYA1 (3), GATA3 (2), HNF1B (6), PAX2 (5), RET (3), ROBO2 (4), SALL1 (9), SIX2 (1), and SIX5 (1). Furthermore, several mutations previously reported to be disease-causing are most likely benign variants. Thus, in a large cohort over 6% of families with isolated CAKUT are caused by a mutation in 12 of 17 dominant CAKUT genes. Our report represents one of the most in-depth diagnostic studies of monogenic causes of isolated CAKUT in children.


Subject(s)
Genes, Dominant , Mutation , Vesico-Ureteral Reflux/genetics , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Genetic Testing/methods , Heredity , Heterozygote , Humans , Male , Pedigree , Phenotype , Predictive Value of Tests , Risk Factors , Urogenital Abnormalities
18.
Kidney Int ; 85(6): 1310-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24152966

ABSTRACT

Congenital abnormalities of the kidney and urinary tract (CAKUT) account for approximately half of children with chronic kidney disease and they are the most frequent cause of end-stage renal disease in children in the US. However, its genetic etiology remains mostly elusive. VACTERL association is a rare disorder that involves congenital abnormalities in multiple organs including the kidney and urinary tract in up to 60% of the cases. By homozygosity mapping and whole-exome resequencing combined with high-throughput mutation analysis by array-based multiplex PCR and next-generation sequencing, we identified recessive mutations in the gene TNF receptor-associated protein 1 (TRAP1) in two families with isolated CAKUT and three families with VACTERL association. TRAP1 is a heat-shock protein 90-related mitochondrial chaperone possibly involved in antiapoptotic and endoplasmic reticulum stress signaling. Trap1 is expressed in renal epithelia of developing mouse kidney E13.5 and in the kidney of adult rats, most prominently in proximal tubules and in thick medullary ascending limbs of Henle's loop. Thus, we identified mutations in TRAP1 as highly likely causing CAKUT or VACTERL association with CAKUT.


Subject(s)
Anal Canal/abnormalities , DNA Mutational Analysis , Esophagus/abnormalities , Exosomes , Genetic Testing , HSP90 Heat-Shock Proteins , Heart Defects, Congenital/genetics , Kidney/abnormalities , Limb Deformities, Congenital/genetics , Mutation , Spine/abnormalities , Trachea/abnormalities , Vesico-Ureteral Reflux/genetics , Age Factors , Animals , DNA Mutational Analysis/methods , Europe , Female , Gene Expression Regulation, Developmental , Genetic Predisposition to Disease , Genetic Testing/methods , Gestational Age , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Heart Defects, Congenital/diagnosis , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Kidney/embryology , Kidney/metabolism , Limb Deformities, Congenital/diagnosis , Male , Mice , Multiplex Polymerase Chain Reaction , Pedigree , Predictive Value of Tests , Risk Factors , United States , Urogenital Abnormalities , Vesico-Ureteral Reflux/diagnosis
19.
Clin Dysmorphol ; 21(4): 191-195, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22895008

ABSTRACT

The VATER/VACTERL association is typically defined by the presence of at least three of the following congenital malformations: vertebral anomalies, anal atresia, cardiac malformations, tracheo-esophageal fistula, renal anomalies, and limb abnormalities. The identification of 14 twin pairs with an initial diagnosis of VATER/VACTERL association at our clinical centers led to the performance of a classical twin study. This involved a thorough evaluation of these 14 twin pairs and a further 55 twin pairs identified from a systematic review of the literature. The zygosity, concordance, and malformation status of all 69 twin pairs were evaluated. Twenty-four twin pairs fulfilled the criteria for inclusion in a comparison of the concordance rates between monozygous (MZ) and dizygous (DZ) twin pairs. The pairwise concordance rates were 15% [95% confidence interval (CI) 4-42%] for MZ and 18% (95% CI 5-48%) for DZ twin pairs (P=0.53). The probandwise concordance rates were 27% (95% CI 11-52%) for MZ and 31% (95% CI 13-58%) for DZ twin pairs (P=0.40). Although based on a limited number of twin pairs, the findings of the present study are consistent with the low number of familial cases reported to date, and suggest that the role of inherited genetic factors in the majority of VATER/VACTERL cases is limited.


Subject(s)
Anus, Imperforate/pathology , Diseases in Twins/pathology , Heart Defects, Congenital/pathology , Esophagus/abnormalities , Esophagus/pathology , Humans , Radius/abnormalities , Radius/pathology , Spine/abnormalities , Spine/pathology , Trachea/abnormalities , Trachea/pathology , Twins, Dizygotic , Twins, Monozygotic
20.
Cell ; 145(4): 513-28, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21565611

ABSTRACT

Nephronophthisis (NPHP), Joubert (JBTS), and Meckel-Gruber (MKS) syndromes are autosomal-recessive ciliopathies presenting with cystic kidneys, retinal degeneration, and cerebellar/neural tube malformation. Whether defects in kidney, retinal, or neural disease primarily involve ciliary, Hedgehog, or cell polarity pathways remains unclear. Using high-confidence proteomics, we identified 850 interactors copurifying with nine NPHP/JBTS/MKS proteins and discovered three connected modules: "NPHP1-4-8" functioning at the apical surface, "NPHP5-6" at centrosomes, and "MKS" linked to Hedgehog signaling. Assays for ciliogenesis and epithelial morphogenesis in 3D renal cultures link renal cystic disease to apical organization defects, whereas ciliary and Hedgehog pathway defects lead to retinal or neural deficits. Using 38 interactors as candidates, linkage and sequencing analysis of 250 patients identified ATXN10 and TCTN2 as new NPHP-JBTS genes, and our Tctn2 mouse knockout shows neural tube and Hedgehog signaling defects. Our study further illustrates the power of linking proteomic networks and human genetics to uncover critical disease pathways.


Subject(s)
Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Signal Transduction , Animals , Ataxin-10 , Centrosome/metabolism , Cilia/metabolism , Ciliary Motility Disorders/genetics , Encephalocele/genetics , Hedgehog Proteins/metabolism , Humans , Kidney Diseases, Cystic/metabolism , Mice , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Polycystic Kidney Diseases/genetics , Retinitis Pigmentosa , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL
...