Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 510
Filtrar
1.
Genes (Basel) ; 15(6)2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38927638

RESUMEN

Approximately half of the cases of chronic kidney disease (CKD) in childhood are caused by congenital anomalies of the kidney and urinary tract (CAKUT). Specific genes were identified as having significant importance in regard to the underlying genetic factors responsible for the CAKUT phenotype, and in our research, we focused on analyzing and comparing the expression levels of ectodysplasin A2 receptor (EDA2R), protocadherin9 (PCDH9), and TNF receptor-associated factor 7 (TRAF7) proteins in the cortex and medulla of healthy control kidneys during developmental phases 2, 3, and 4. We also performed an analysis of the area percentages of the mentioned proteins in the cortical and medullary sections of healthy embryonic and fetal kidneys compared to those affected by CAKUT, including duplex kidneys (DK), horseshoe kidneys (HK), hypoplastic kidneys (HYP), and dysplastic kidneys (DYS). We found that the CAKUT candidate gene proteins EDA2R, PCDH9, and TRAF7 are all expressed during normal human kidney development stages. In DYS, the expression of EDA2R was higher than in normal kidneys, likely due to EDA2R's role in apoptosis, which was upregulated in specific cases and could possibly contribute to the formation of DYS. The expression of PCDH9 was lower in HK, which can be attributed to the possible role of PCDH9 in cell migration suppression. Decreased PCDH9 expression is linked to increased cell migration, potentially contributing to the development of HK. The level of TRAF7 expression was reduced in all examined kidney disorders compared to normal kidneys, suggesting that this reduction might be attributed to the crucial role of TRAF7 in the formation of endothelium and ciliogenesis, both of which are essential for normal kidney development. Further research is required to ascertain the function of these proteins in both the typical development of the kidney and in CAKUT.


Asunto(s)
Cadherinas , Riñón , Anomalías Urogenitales , Reflujo Vesicoureteral , Humanos , Riñón/metabolismo , Riñón/anomalías , Riñón/crecimiento & desarrollo , Riñón/embriología , Reflujo Vesicoureteral/genética , Reflujo Vesicoureteral/patología , Cadherinas/genética , Cadherinas/metabolismo , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología , Protocadherinas , Sistema Urinario/anomalías , Sistema Urinario/metabolismo , Regulación del Desarrollo de la Expresión Génica
2.
Hum Genomics ; 18(1): 41, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38654324

RESUMEN

BACKGROUND: Congenital anomalies of the kidney and urinary tract (CAKUT) are prevalent birth defects. Although pathogenic CAKUT genes are known, they are insufficient to reveal the causes for all patients. Our previous studies indicated GEN1 as a pathogenic gene of CAKUT in mice, and this study further investigated the correlation between GEN1 and human CAKUT. METHODS: In this study, DNA from 910 individuals with CAKUT was collected; 26 GEN1 rare variants were identified, and two GEN1 (missense) variants in a non-CAKUT group were found. Mainly due to the stability results of the predicted mutant on the website, in vitro, 10 variants (eight CAKUT, two non-CAKUT) were selected to verify mutant protein stability. In addition, mainly based on the division of the mutation site located in the functional region of the GEN1 protein, 8 variants (six CAKUT, two non-CAKUT) were selected to verify enzymatic hydrolysis, and the splice variant GEN1 (c.1071 + 3(IVS10) A > G) was selected to verify shear ability. Based on the results of in vitro experiments and higher frequency, three sites with the most significant functional change were selected to build mouse models. RESULTS: Protein stability changed in six variants in the CAKUT group. Based on electrophoretic mobility shift assay of eight variants (six CAKUT, two non-CAKUT), the enzymatic hydrolysis and DNA-binding abilities of mutant proteins were impaired in the CAKUT group. The most serious functional damage was observed in the Gen1 variant that produced a truncated protein. A mini-gene splicing assay showed that the variant GEN1 (c.1071 + 3(IVS10) A > G) in the CAKUT group significantly affected splicing function. An abnormal exon10 was detected in the mini-gene splicing assay. Point-mutant mouse strains were constructed (Gen1: c.1068 + 3 A > G, p.R400X, and p.T105R) based on the variant frequency in the CAKUT group and functional impairment in vitro study and CAKUT phenotypes were replicated in each. CONCLUSION: Overall, our findings indicated GEN1 as a risk factor for human CAKUT.


Asunto(s)
Anomalías Urogenitales , Reflujo Vesicoureteral , Animales , Femenino , Humanos , Masculino , Ratones , Predisposición Genética a la Enfermedad , Riñón/anomalías , Riñón/patología , Riñón/metabolismo , Mutación/genética , Estabilidad Proteica , Factores de Riesgo , Sistema Urinario/anomalías , Sistema Urinario/patología , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología , Reflujo Vesicoureteral/genética , Reflujo Vesicoureteral/patología
3.
Biochem Biophys Res Commun ; 714: 149959, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38657443

RESUMEN

Gestational diabetes mellitus (GDM) presents a substantial population health concern. Previous studies have revealed that GDM can ultimately influence nephron endowment. In this study, we established a GDM mouse model to investigate the embryological alterations and molecular mechanisms underlying the development of congenital anomalies of the kidney and urinary tract (CAKUT) affected by GDM. Our study highlights that GDM could contribute to the manifestation of CAKUT, with prevalent phenotypes characterized by isolated hydronephrosis and duplex kidney complicated with hydronephrosis in mice. Ectopic ureteric buds (UBs) and extended length of common nephric ducts (CNDs) were noted in the metanephric development stage. The expression of Ret and downstream p-ERK activity were enhanced in UBs, which indicated the alteration of RET/MAPK/ERK pathway may be one of the mechanisms contributing to the increased occurrence of CAKUT associated with GDM.


Asunto(s)
Diabetes Gestacional , Sistema de Señalización de MAP Quinasas , Proteínas Proto-Oncogénicas c-ret , Anomalías Urogenitales , Reflujo Vesicoureteral , Animales , Femenino , Ratones , Embarazo , Diabetes Gestacional/metabolismo , Riñón/anomalías , Riñón/metabolismo , Riñón/embriología , Proteínas Proto-Oncogénicas c-ret/metabolismo , Proteínas Proto-Oncogénicas c-ret/genética , Sistema Urinario/anomalías , Sistema Urinario/embriología , Anomalías Urogenitales/etiología , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología
4.
Clin Genet ; 106(1): 102-108, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38558253

RESUMEN

Pathogenic germline variants in the FOXL2 gene are associated with Blepharophimosis, Ptosis, and Epicanthus Inversus syndrome (BPES) in humans, an autosomal dominant condition. Two forms of BPES have emerged: (i) type I (BPES-I), characterized by ocular signs and primary ovarian failure (POI), and (ii) type II (BPES-II) with no systemic associations. This study aimed to compare the distribution of FOXL2 variants in idiopathic POI/DOR (diminished ovarian reserve) and both types of BPES, and to determine the involvement of FOXL2 in non-syndromic forms of POI/DOR. We studied the whole coding region of the FOXL2 gene using next-generation sequencing in 1282 patients with non-syndromic POI/DOR. Each identified FOXL2 variant was compared to its frequency in the general population, considering ethnicity. Screening of the entire coding region of the FOXL2 gene allowed us to identify 10 different variants, including nine missense variants. Of the patients with POI/DOR, 14 (1%) carried a FOXL2 variant. Significantly, six out of nine missense variants (67%) were overrepresented in our POI/DOR cohort compared to the general or specific ethnic subgroups. Our findings strongly suggest that five rare missense variants, mainly located in the C-terminal region of FOXL2 are high-risk factors for non-syndromic POI/DOR, though FOXL2 gene implication accounts for approximately 0.54% of non-syndromic POI/DOR cases. These results support the implementation of routine genetic screening for patients with POI/DOR in clinical settings.


Asunto(s)
Blefarofimosis , Proteína Forkhead Box L2 , Mutación Missense , Insuficiencia Ovárica Primaria , Humanos , Proteína Forkhead Box L2/genética , Femenino , Insuficiencia Ovárica Primaria/genética , Mutación Missense/genética , Blefarofimosis/genética , Adulto , Secuenciación de Nucleótidos de Alto Rendimiento , Predisposición Genética a la Enfermedad , Anomalías Cutáneas/genética , Anomalías Urogenitales/genética , Factores de Transcripción Forkhead/genética , Fenotipo
5.
Pediatr Nephrol ; 39(8): 2351-2353, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38467926

RESUMEN

Renal coloboma syndrome (RCS) and dominant optic atrophy are mainly caused by heterozygous mutations in PAX2 and OPA1, respectively. We describe a patient with digenic mutations in PAX2 and OPA1. A female infant was born without perinatal abnormalities. Magnetic resonance imaging at 4 months of age showed bilateral microphthalmia and optic nerve hypoplasia. Appropriate body size was present at 2 years of age, and mental development was favorable. Color fundus photography revealed severe retinal atrophy in both eyes. Electroretinography showed slight responses in the right eye, but no responses in the left eye, suggesting a high risk of blindness. Urinalysis results were normal, creatinine-based estimated glomerular filtration rate was 63.5 mL/min/1.73 m2, and ultrasonography showed bilateral hypoplastic kidneys. Whole exome sequencing revealed de novo frameshift mutations in PAX2 and OPA1. Both variants were classified as pathogenic (PVS1, PS2, PM2) based on the guidelines from the American College of Medical Genetics and Genomics (ACMG). Genetic testing for ocular diseases should be considered for patients with suspected RCS and a high risk of total blindness.


Asunto(s)
Coloboma , GTP Fosfohidrolasas , Factor de Transcripción PAX2 , Reflujo Vesicoureteral , Humanos , Femenino , Factor de Transcripción PAX2/genética , GTP Fosfohidrolasas/genética , Coloboma/genética , Coloboma/diagnóstico , Reflujo Vesicoureteral/genética , Reflujo Vesicoureteral/diagnóstico , Atrofia Óptica Autosómica Dominante/genética , Atrofia Óptica Autosómica Dominante/diagnóstico , Anomalías Urogenitales/genética , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/complicaciones , Mutación del Sistema de Lectura , Secuenciación del Exoma , Lactante , Preescolar , Mutación , Insuficiencia Renal
6.
Am J Med Genet A ; 194(7): e63582, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38450833

RESUMEN

We present three new and six published infants with overlapping features of LUMBAR syndrome (lower body hemangioma, urogenital anomalies, spinal cord malformations, bony deformities, anorectal/arterial anomalies and renal anomalies) and OEIS complex (omphalocele, exstrophy, imperforate anus, and spinal defects), also known as cloacal exstrophy. OEIS is included under the recently proposed umbrella coined recurrent constellations of embryonic malformations (RCEMs). The RCEMs represent a phenotypically overlapping spectrum of rare disorders of caudal dysgenesis with unknown cause but likely shared pathogenesis. It has recently been proposed that LUMBAR be considered an RCEM. This report of infants with combined features of OEIS and LUMBAR is the first to demonstrate an overlap between LUMBAR and another RCEM, which supports LUMBAR's inclusion within the RCEM spectrum.


Asunto(s)
Anomalías Múltiples , Ano Imperforado , Humanos , Ano Imperforado/genética , Ano Imperforado/patología , Ano Imperforado/diagnóstico , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Anomalías Múltiples/diagnóstico , Femenino , Masculino , Recién Nacido , Anomalías Urogenitales/genética , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/patología , Hernia Umbilical/diagnóstico , Hernia Umbilical/patología , Lactante , Síndrome , Cloaca/anomalías , Cloaca/patología , Hemangioma/patología , Hemangioma/diagnóstico , Hemangioma/genética , Fenotipo , Columna Vertebral/anomalías , Columna Vertebral/patología , Columna Vertebral/diagnóstico por imagen , Escoliosis
7.
Eur J Pediatr ; 183(5): 2257-2272, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38411716

RESUMEN

Patients with Aarskog-Scott syndrome (AAS) have short stature, facial anomalies, skeletal deformities, and genitourinary malformations. FYVE, RhoGEF, and PH domain-containing 1 (FGD1) is the only known causative gene of AAS. However, the diagnosis of AAS remains difficult, and specific treatments are still absent. Patients suspected with AAS were recruited, and clinical information was collected. Genetic testing and functional analysis were carried out for the diagnosis. By literature review, we summarized the clinical and genetic characteristics of FGD1-related AAS and analyzed the genotype-phenotype correlation. Five patients were recruited, and four novel FGD1 variants were identified. The diagnosis of AAS was confirmed by genetic analysis and functional study. Three patients treated with growth hormone showed improved heights during the follow-up period. By literature review, clinical features of AAS patients with FGD1 variants were summarized. Regarding FGD1 variations, substitutions were the most common form, and among them, missense variants were the most frequent. Moreover, we found patients with drastic variants showed higher incidences of foot and genitourinary malformations. Missense variants in DH domain were related to a lower incidence of cryptorchidism.   Conclusion: We reported four novel pathogenic FGD1 variations in AAS patients and confirmed the efficacy and safety of growth hormone treatment in FGD1-related AAS patients with growth hormone deficiency. Additionally, our literature review suggested the crucial role of DH domain in FGD1 function. What is Known: • Aarskog-Scott syndrome is a rare genetic disease, and the only known cause is the variant in FGD1 gene. The typical clinical manifestations of AAS include facial, skeletal, and urogenital deformities and short stature. What is New: • We reported four novel FGD1 variants and reported the treatment of growth hormone in FGD1-related AAS patients. Our genotype-phenotype correlation analysis suggested the crucial role of DH domain in FGD1 function.


Asunto(s)
Anomalías Múltiples , Cara , Enfermedades Genéticas Ligadas al Cromosoma X , Genitales Masculinos , Factores de Intercambio de Guanina Nucleótido , Niño , Preescolar , Femenino , Humanos , Masculino , Anomalías Múltiples/genética , Anomalías Múltiples/diagnóstico , Enanismo/genética , Enanismo/diagnóstico , Enanismo/tratamiento farmacológico , Cara/anomalías , Estudios de Asociación Genética , Genitales Masculinos/anomalías , Factores de Intercambio de Guanina Nucleótido/genética , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/diagnóstico , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/diagnóstico , Fenotipo , Dermatosis del Cuero Cabelludo/genética , Dermatosis del Cuero Cabelludo/diagnóstico , Dermatosis del Cuero Cabelludo/tratamiento farmacológico , Dermatosis del Cuero Cabelludo/congénito , Anomalías Urogenitales/genética , Anomalías Urogenitales/diagnóstico
8.
Prenat Diagn ; 44(3): 343-351, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38285371

RESUMEN

OBJECTIVE: GREB1L has been linked prenatally to Potter's sequence, as well as less severe anomalies of the kidney, uterus, inner ear, and heart. The full phenotypic spectrum is unknown. The purpose of this study was to characterize known and novel pre- and postnatal phenotypes associated with GREB1L. METHODS: We solicited cases from the Fetal Sequencing Consortium, screened a population-based genomic database, and conducted a comprehensive literature search to identify disease cases associated with GREB1L. We present a detailed phenotypic spectrum and molecular changes. RESULTS: One hundred twenty-seven individuals with 51 unique pathogenic or likely pathogenic GREB1L variants were identified. 24 (47%) variants were associated with isolated kidney anomalies, 19 (37%) with anomalies of multiple systems, including one case of hypoplastic left heart syndrome, five (10%) with isolated sensorineural hearing loss, two (4%) with isolated uterine agenesis; and one (2%) with isolated tetralogy of Fallot. CONCLUSION: GREB1L may cause complex congenital heart disease (CHD) in humans. Clinicians should consider GREB1L testing in the setting of CHD, and cardiac screening in the setting of GREB1L variants.


Asunto(s)
Cardiopatías Congénitas , Enfermedades Renales , Anomalías Urogenitales , Femenino , Humanos , Cardiopatías Congénitas/epidemiología , Cardiopatías Congénitas/genética , Riñón/anomalías , Enfermedades Renales/congénito , Proteínas de Neoplasias/genética , Anomalías Urogenitales/genética
9.
Int J Gynaecol Obstet ; 166(1): 353-359, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38189110

RESUMEN

OBJECTIVE: Duplex kidney is a relatively frequent form of urinary system abnormality. This study aimed to elucidate the value of chromosomal microarray analysis (CMA) and whole exome sequencing (WES) for duplex kidney and the perinatal outcomes of duplex kidney fetuses. METHODS: This retrospective cohort study included 63 patients with duplex kidney diagnosed using antenatal ultrasound between August 2013 and January 2023. We reviewed the clinical characteristics, genetic test results, and pregnancy outcomes of the patients. RESULTS: Among the 63 cases based on the inclusion criteria, the CMA detected seven (11.1%) clinically significant variants and nine variants of uncertain significance (VUS), and the pathogenic/likely pathogenic (P/LP) copy number variations (CNVs) in the recurrent region that were associated with prenatal duplex kidney included 17q12, 17p13.3, and 22q11.2. No significant disparity was observed in the CMA detection rate between the unilateral and bilateral groups, or between the isolated and non-isolated groups. WES identified three (50%) P/LP single-gene variants in six fetuses with duplex kidney. We detected the following pathogenic genes in the duplex kidney fetuses: KMT2D, SMPD4, and FANCI. Pregnancy termination in cases where clinically significant variants were detected by genetic testing was different in statistical significance from that in cases with negative results (9/10, 90.0% vs 8/48, 16.7%, P < 0.001). CONCLUSION: This study elucidated the value of CMA and WES for fetal duplex kidney, proving that CMA and WES may be useful tools in prenatal diagnosis and genetic counseling.


Asunto(s)
Secuenciación del Exoma , Riñón , Ultrasonografía Prenatal , Humanos , Femenino , Embarazo , Estudios Retrospectivos , Adulto , Riñón/anomalías , Riñón/diagnóstico por imagen , Resultado del Embarazo , Diagnóstico Prenatal/métodos , Variaciones en el Número de Copia de ADN , Anomalías Urogenitales/genética , Anomalías Urogenitales/diagnóstico por imagen , Anomalías Urogenitales/diagnóstico , Pruebas Genéticas/métodos , Análisis por Micromatrices
10.
Prenat Diagn ; 44(2): 205-221, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38180355

RESUMEN

OBJECTIVE: The goal of this study was to review and analyze the medical literature for cases of prenatal and/or postnatally diagnosed bilateral renal agenesis (BRA) and create a comprehensive summary of the genetic etiologies known to be associated with this condition. METHODS: A literature search was conducted as a scoping review employing Online Mendeliain Inheritance in Man, PubMed, and Cochrane to identify cases of BRA with known underlying genetic (chromosomal vs. single gene) etiologies and those described in syndromes without any known genetic etiology. The cases were further categorized as isolated versus non-isolated, describing additional findings reported prenatally, postnatally, and postmortem. Inheritance pattern was also documented when appropriate in addition to the reported timing of diagnosis and sex. RESULTS: We identified six cytogenetic abnormalities and 21 genes responsible for 20 single gene disorders associated with BRA. Five genes have been reported to associate with BRA without other renal anomalies; sixteen others associate with both BRA as well as unilateral renal agenesis. Six clinically recognized syndromes/associations were identified with an unknown underlying genetic etiology. Genetic etiologies of BRA are often phenotypically expressed as other urogenital anomalies as well as complex multi-system syndromes. CONCLUSION: Multiple genetic etiologies of BRA have been described, including cytogenetic abnormalities and monogenic syndromes. The current era of the utilization of exome and genome-wide sequencing is likely to significantly expand our understanding of the underlying genetic architecture of BRA.


Asunto(s)
Anomalías Congénitas , Enfermedades Renales , Enfermedades Renales/congénito , Riñón/anomalías , Anomalías Urogenitales , Embarazo , Femenino , Humanos , Enfermedades Renales/genética , Anomalías Urogenitales/genética , Aberraciones Cromosómicas , Síndrome
11.
Am J Med Genet A ; 194(5): e63526, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38192228

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are estimated to be responsible for 20%-50% of congenital anomalies and are also a leading etiology of early-onset renal disease. Primary CAKUT are caused by genetic factors that impair proper in-utero genitourinary tract development and secondary CAKUT result from the influence of environmental factors. The CHRNA3 gene, which encodes the Alpha-3 subunit of the nicotinic acetylcholine receptor, is hypothesized to be associated with Megacystis-microcolon-intestinal hyperperistalsis syndrome. More recently, pathogenic variants in CHRNA3 have been identified in individuals with CAKUT as well as individuals with panautonomic failure. Here we present a patient with neurogenic bladder, vesicoureteral reflux, mydriasis, and gastrointestinal dysmotility found to have novel compound heterozygous variants in CHRNA3. These findings support the consideration of CHRNA3 disruption in the differential for CAKUT with dysautonomia and gastrointestinal dysmotility.


Asunto(s)
Enfermedades del Sistema Nervioso Autónomo , Receptores Nicotínicos , Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Humanos , Vejiga Urinaria , Riñón/anomalías , Reflujo Vesicoureteral/genética , Anomalías Urogenitales/genética , Enfermedades del Sistema Nervioso Autónomo/patología , Receptores Nicotínicos/genética
12.
Pediatr Nephrol ; 39(2): 357-369, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-37468646

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are among the most common birth defects worldwide and a major cause of kidney failure in children. Extra-renal manifestations are also common. This study reviewed diseases associated with the Genomics England CAKUT-associated gene panel for ocular anomalies. In addition, each gene was examined for expression in the human retina and an ocular phenotype in mouse models using the Human Protein Atlas and Mouse Genome Informatics databases, respectively. Thirty-four (54%) of the 63 CAKUT-associated genes (55 'green' and 8 'amber') had a reported ocular phenotype. Five of the 6 most common CAKUT-associated genes (PAX2, EYA1, SALL1, GATA3, PBX1) that represent 30% of all diagnoses had ocular features. The ocular abnormalities found with most CAKUT-associated genes and with five of the six commonest were coloboma, microphthalmia, optic disc anomalies, refraction errors (astigmatism, myopia, and hypermetropia), and cataract. Seven of the CAKUT-associated genes studied (11%) had no reported ocular features but were expressed in the human retina or had an ocular phenotype in a mouse model, which suggested further possibly-unrecognised abnormalities. About one third of CAKUT-associated genes (18, 29%) had no ocular associations and were not expressed in the retina, and the corresponding mouse models had no ocular phenotype. Ocular abnormalities in individuals with CAKUT suggest a genetic basis for the disease and sometimes indicate the affected gene. Individuals with CAKUT often have ocular abnormalities and may require an ophthalmic review, monitoring, and treatment to preserve vision.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Niño , Ratones , Animales , Humanos , Riñón/metabolismo , Sistema Urinario/anomalías , Reflujo Vesicoureteral/genética , Reflujo Vesicoureteral/diagnóstico , Anomalías Urogenitales/genética , Anomalías Urogenitales/diagnóstico
13.
Kidney Int ; 105(4): 844-864, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38154558

RESUMEN

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


Asunto(s)
Estructuras Embrionarias , Factores de Transcripción Forkhead , Enfermedades Renales , Riñón , Nefronas , Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Adulto , Animales , Humanos , Ratones , Estudio de Asociación del Genoma Completo , Riñón/anomalías , Riñón/embriología , Enfermedades Renales/genética , Ratones Noqueados , Nefronas/embriología , Factores de Transcripción/genética , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/metabolismo
14.
Genet Med ; 25(12): 100983, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37746849

RESUMEN

PURPOSE: Previous work identified rare variants in DSTYK associated with human congenital anomalies of the kidney and urinary tract (CAKUT). Here, we present a series of mouse and human studies to clarify the association, penetrance, and expressivity of DSTYK variants. METHODS: We phenotypically characterized Dstyk knockout mice of 3 separate inbred backgrounds and re-analyzed the original family segregating the DSTYK c.654+1G>A splice-site variant (referred to as "SSV" below). DSTYK loss of function (LOF) and SSVs were annotated in individuals with CAKUT, epilepsy, or amyotrophic lateral sclerosis vs controls. A phenome-wide association study analysis was also performed using United Kingdom Biobank (UKBB) data. RESULTS: Results demonstrate ∼20% to 25% penetrance of obstructive uropathy, at least, in C57BL/6J and FVB/NJ Dstyk-/- mice. Phenotypic penetrance increased to ∼40% in C3H/HeJ mutants, with mild-to-moderate severity. Re-analysis of the original family segregating the rare SSV showed low penetrance (43.8%) and no alternative genetic causes for CAKUT. LOF DSTYK variants burden showed significant excess for CAKUT and epilepsy vs controls and an exploratory phenome-wide association study supported association with neurological disorders. CONCLUSION: These data support causality for DSTYK LOF variants and highlights the need for large-scale sequencing studies (here >200,000 cases) to accurately assess causality for genes and variants to lowly penetrant traits with common population prevalence.


Asunto(s)
Epilepsia , Sistema Urinario , Anomalías Urogenitales , Animales , Ratones , Humanos , Penetrancia , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Anomalías Urogenitales/genética , Riñón/anomalías , Factores de Riesgo , Epilepsia/genética , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética
15.
Clin Genet ; 104(6): 679-685, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37468454

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) is the leading cause of chronic kidney disease in the first three decades of life. Until now, more than 180 monogenic causes of isolated or syndromic CAKUT have been described. In addition, copy number variants (CNV) have also been implicated, however, all of these causative factors only explain a small fraction of patients with CAKUT, suggesting that additional yet-to-be-discovered novel genes are present. Herein, we report three siblings (two of them are monozygotic twin) of a consanguineous family with CAKUT. Whole-exome sequencing identified a homozygous variant in TBC1D31. Three dimensional protein modeling as well as molecular dynamics simulations predicted it as pathogenic. We therefore showed for the first time an association between a homozygous TBC1D31 variant with CAKUT in humans, expanding its genetic spectrum.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Humanos , Consanguinidad , Riñón/anomalías , Anomalías Urogenitales/genética
16.
Am J Med Genet A ; 191(8): 2083-2091, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37213061

RESUMEN

Neurogenic bladder is caused by disruption of neuronal pathways regulating bladder relaxation and contraction. In severe cases, neurogenic bladder can lead to vesicoureteral reflux, hydroureter, and chronic kidney disease. These complications overlap with manifestations of congenital anomalies of the kidney and urinary tract (CAKUT). To identify novel monogenic causes of neurogenic bladder, we applied exome sequencing (ES) to our cohort of families with CAKUT. By ES, we have identified a homozygous missense variant (p.Gln184Arg) in CHRM5 (cholinergic receptor, muscarinic, 5) in a patient with neurogenic bladder and secondary complications of CAKUT. CHRM5 codes for a seven transmembrane-spanning G-protein-coupled muscarinic acetylcholine receptor. CHRM5 is shown to be expressed in murine and human bladder walls and is reported to cause bladder overactivity in Chrm5 knockout mice. We investigated CHRM5 as a potential novel candidate gene for neurogenic bladder with secondary complications of CAKUT. CHRM5 is similar to the cholinergic bladder neuron receptor CHRNA3, which Mann et al. published as the first monogenic cause of neurogenic bladder. However, functional in vitro studies did not reveal evidence to strengthen the status as a candidate gene. Discovering additional families with CHRM5 variants could help to further assess the genes' candidate status.


Asunto(s)
Vejiga Urinaria Neurogénica , Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Humanos , Ratones , Animales , Vejiga Urinaria Neurogénica/genética , Anomalías Urogenitales/genética , Reflujo Vesicoureteral/genética , Riñón/anomalías , Ratones Noqueados
17.
Pediatr Nephrol ; 38(12): 3963-3973, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-36867265

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) result from disruptions in normal kidney and urinary tract development during fetal life and collectively represent the most common cause of kidney failure in children worldwide. The antenatal determinants of CAKUT are diverse and include mutations in genes responsible for normal nephrogenesis, alterations in maternal and fetal environments, and obstruction within the normal developing urinary tract. The resultant clinical phenotypes are complex and depend on the timing of the insult, the penetrance of underlying gene mutations, and the severity and timing of obstruction related to the sequence of normal kidney development. Consequently, there is a broad spectrum of outcomes for children born with CAKUT. In this review, we explore the most common forms of CAKUT and those most likely to develop long-term complications of their associated kidney malformations. We discuss the relevant outcomes for the different forms of CAKUT and what is known about clinical characteristics across the CAKUT spectrum that are risk factors of long-term kidney injury and disease progression.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Niño , Femenino , Humanos , Embarazo , Riñón/anomalías , Sistema Urinario/anomalías , Anomalías Urogenitales/complicaciones , Anomalías Urogenitales/epidemiología , Anomalías Urogenitales/genética , Factores de Riesgo , Progresión de la Enfermedad
18.
Eur J Hum Genet ; 31(6): 674-680, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36922632

RESUMEN

Individuals with congenital anomalies of the kidney and urinary tract (CAKUT) show a broad spectrum of malformations. CAKUT can occur in an isolated fashion or as part of a syndromic disorder and can lead to end-stage kidney failure. A monogenic cause can be identified in ~12% of affected individuals. This study investigated a single-center CAKUT cohort analyzed by exome sequencing (ES). Emphasis was placed on the question whether diagnostic yield differs between certain CAKUT phenotypes (e.g., bilateral kidney affection, unilateral kidney affection or only urinary tract affection). 86 unrelated individuals with CAKUT were categorized according to their phenotype and analyzed by ES to identify a monogenic cause. Prioritized variants were rated according to the recommendations of the American College of Medical Genetics and Genomics and the Association for Clinical Genomic Science. Diagnostic yields of different phenotypic categories were compared. Clinical data were collected using a standardized questionnaire. In the study cohort, 7/86 individuals had a (likely) pathogenic variant in the genes PAX2, PBX1, EYA1, or SALL1. Additionally, in one individual, a 17q12 deletion syndrome (including HNF1B) was detected. 64 individuals had a kidney affection, which was bilateral in 36. All solved cases (8/86, 9%) had bilateral kidney affection (diagnostic yield in subcohort: 8/36, 22%). Although the diagnostic yield in CAKUT cohorts is low, our single-center experience argues, that, in individuals with bilateral kidney affection, monogenic burden is higher than in those with unilateral kidney or only urinary tract affection.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Reflujo Vesicoureteral , Humanos , Secuenciación del Exoma , Riñón/anomalías , Sistema Urinario/anomalías , Reflujo Vesicoureteral/genética , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/genética , Anomalías Urogenitales/patología
19.
Mol Med Rep ; 27(3)2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36734258

RESUMEN

The HOXA genes cluster plays a key role in embryologic development. Mutations in HOXA genes have been linked to different human phenotypes, including developmental delay, limb anomalies, and urogenital malformations. The present study reported a clinical and genetic investigation of a female patient with polymalformative syndrome including left arm agenesis, bicornuate uterus and bicuspid aortic valve. Using whole exome sequencing, two heterozygous missense variants were identified. Of these, one was a novel variant in the HOXA13 gene [p.(Tyr290Ser)] and the second a heterozygous variant in the HOXA9 gene [p.(Ala102Pro)]. To the best of our knowledge, this is the first association of HOXA9/HOXA13 point mutations linked to a syndromic case. In conclusion, the present study suggested that the phenotypic spectrum of vertebral anomalies, anal atresia, cardiac defects, tracheo­esophageal fistula, renal anomalies and limb abnormalities/hand­foot­genital syndrome may be attributable to the combination of different HOXA variants, particularly in patients with a severe clinical presentation. The current report contributed as well to the molecular understanding of HOXA genes­related phenotypes via the identification of novel variant and genes associations.


Asunto(s)
Anomalías Múltiples , Genes Homeobox , Anomalías Urogenitales , Femenino , Humanos , Anomalías Múltiples/genética , Mutación , Fenotipo , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/genética
20.
Development ; 150(3)2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36789910

RESUMEN

Frizzled 2 (FZD2) is a transmembrane Wnt receptor. We previously identified a pathogenic human FZD2 variant in individuals with FZD2-associated autosomal dominant Robinow syndrome. The variant encoded a protein with a premature stop and loss of 17 amino acids, including a region of the consensus dishevelled-binding sequence. To model this variant, we used zygote microinjection and i-GONAD-based CRISPR/Cas9-mediated genome editing to generate a mouse allelic series. Embryos mosaic for humanized Fzd2W553* knock-in exhibited cleft palate and shortened limbs, consistent with patient phenotypes. We also generated two germline mouse alleles with small deletions: Fzd2D3 and Fzd2D4. Homozygotes for each allele exhibit a highly penetrant cleft palate phenotype, shortened limbs compared with wild type and perinatal lethality. Fzd2D4 craniofacial tissues indicated decreased canonical Wnt signaling. In utero treatment with IIIC3a (a DKK inhibitor) normalized the limb lengths in Fzd2D4 homozygotes. The in vivo replication represents an approach for further investigating the mechanism of FZD2 phenotypes and demonstrates the utility of CRISPR knock-in mice as a tool for investigating the pathogenicity of human genetic variants. We also present evidence for a potential therapeutic intervention.


Asunto(s)
Fisura del Paladar , Enanismo , Deformidades Congénitas de las Extremidades , Anomalías Urogenitales , Animales , Humanos , Ratones , Fisura del Paladar/genética , Enanismo/genética , Deformidades Congénitas de las Extremidades/genética , Anomalías Urogenitales/genética , Vía de Señalización Wnt/genética , Modelos Animales de Enfermedad , Receptores Frizzled/genética , Técnicas de Sustitución del Gen
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA