Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Genet Med ; 22(8): 1374-1383, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32398770

RESUMEN

PURPOSE: Autosomal dominant polycystic kidney disease (ADPKD) represents the most common hereditary nephropathy. Despite growing evidence for genetic heterogeneity, ADPKD diagnosis is still primarily based upon clinical imaging criteria established before discovery of additional PKD genes. This study aimed at assessing the diagnostic value of genetic verification in clinical ADPKD. METHODS: In this prospective, diagnostic trial, 100 families with clinically diagnosed ADPKD were analyzed by PKD gene panel and multiplex ligation-dependent probe amplification (MLPA); exome sequencing (ES) was performed in panel/MLPA-negative families. RESULTS: Diagnostic PKD1/2 variants were identified in 81 families (81%), 70 of which in PKD1 and 11 in PKD2. PKD1 variants of unknown significance were detected in another 9 families (9%). Renal survival was significantly worse upon PKD1 truncation versus nontruncation and PKD2 alteration. Ten percent of the cohort were PKD1/2-negative, revealing alternative genetic diagnoses such as autosomal recessive PKD, Birt-Hogg-Dubé syndrome, and ALG9-associated PKD. In addition, among unsolved cases, ES yielded potential novel PKD candidates. CONCLUSION: By illustrating vast genetic heterogeneity, this study demonstrates the value of genetic testing in a real-world PKD cohort by diagnostic verification, falsification, and disease prediction. In the era of specific treatment for fast progressive ADPKD, genetic confirmation should form the basis of personalized patient care.


Asunto(s)
Riñón Poliquístico Autosómico Dominante , Pruebas Genéticas , Humanos , Mutación , Riñón Poliquístico Autosómico Dominante/diagnóstico , Riñón Poliquístico Autosómico Dominante/genética , Estudios Prospectivos , Canales Catiónicos TRPP/genética
2.
Hum Mol Genet ; 22(11): 2177-85, 2013 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-23418306

RESUMEN

Mutations affecting the integrity and function of cilia have been identified in various genes over the last decade accounting for a group of diseases called ciliopathies. Ciliopathies display a broad spectrum of phenotypes ranging from mild manifestations to lethal combinations of multiple severe symptoms and most of them share cystic kidneys as a common feature. Our starting point was a consanguineous pedigree with three affected fetuses showing an early embryonic phenotype with enlarged cystic kidneys, liver and pancreas and developmental heart disease. By genome-wide linkage analysis, we mapped the disease locus to chromosome 17q11 and identified a homozygous nonsense mutation in NEK8/NPHP9 that encodes a kinase involved in ciliary dynamics and cell cycle progression. Missense mutations in NEK8/NPHP9 have been identified in juvenile cystic kidney jck mice and in patients suffering from nephronophthisis (NPH), an autosomal-recessive cystic kidney disease. This work confirmed a complete loss of NEK8 expression in the affected fetuses due to nonsense-mediated decay. In cultured fibroblasts derived from these fetuses, the expression of prominent polycystic kidney disease genes (PKD1 and PKD2) was decreased, whereas the oncogene c-MYC was upregulated, providing potential explanations for the observed renal phenotype. We furthermore linked NEK8 with NPHP3, another NPH protein known to cause a very similar phenotype in case of null mutations. Both proteins interact and activate the Hippo effector TAZ. Taken together, our study demonstrates that NEK8 is essential for organ development and that the complete loss of NEK8 perturbs multiple signalling pathways resulting in a severe early embryonic phenotype.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Síndrome de Dandy-Walker/genética , Síndrome de Dandy-Walker/metabolismo , Regulación de la Expresión Génica , Mutación , Quiste Pancreático/genética , Quiste Pancreático/metabolismo , Proteínas Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Transducción de Señal , Anomalías Múltiples/patología , Línea Celular , Consanguinidad , Síndrome de Dandy-Walker/patología , Femenino , Feto/anomalías , Frecuencia de los Genes , Estudio de Asociación del Genoma Completo , Genotipo , Vía de Señalización Hippo , Humanos , Masculino , Quinasas Relacionadas con NIMA , Quiste Pancreático/patología , Linaje , Polimorfismo de Nucleótido Simple , Unión Proteica , Canales Catiónicos TRPP/genética , Canales Catiónicos TRPP/metabolismo
3.
Pediatr Nephrol ; 29(8): 1451-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24504730

RESUMEN

BACKGROUND: An emerging number of clinically and genetically heterogeneous diseases now collectively termed ciliopathies have been connected to the dysfunction of primary cilia. We describe an 8-year-old girl with a complex phenotype that did not clearly match any familiar syndrome. CASE-DIAGNOSIS/TREATMENT: Hypotonia, facial dysmorphism and retardation were noted shortly after birth. Other features included short stature, mild skeletal anomalies, strabism, deafness, subdural hygroma, hepatosplenomegaly and end-stage renal failure. Renal biopsy revealed tubular atrophy, interstitial fibrosis and segmental glomerulosclerosis. After exclusion of a chromosomal abnormality by array-comparative genomic hybridization (CGH), we performed next-generation sequencing (NGS) using a customized panel that targeted 131 genes known or hypothesized to cause ciliopathies. We identified the novel homozygous WDR19 mutation c.1483G > C (p.Gly495Arg) that affects an evolutionarily highly conserved residue in the intraflagellar transport protein IFT144, is absent from databases and is predicted to be pathogenic by all bioinformatic sources used. CONCLUSION: Mutations in WDR19 encoding the intraflagellar transport component IFT144 have recently been described in single families with the clinically overlapping skeletal ciliopathies Jeune and Sensenbrenner syndromes, combined or isolated nephronophthisis (NPHP) and retinitis pigmentosa (RP) (Senior-Loken syndrome). Our patient emphasizes the usefulness and efficiency of a comprehensive NGS panel approach in patients with unclassified ciliopathies. It further suggests that WDR19 mutations can cause a broad spectrum of ciliopathies that extends to Jeune and Sensenbrenner syndromes, RP and renal NPHP-like phenotypes.


Asunto(s)
Cilios/patología , Enfermedades Renales/genética , Proteínas/genética , Niño , Proteínas del Citoesqueleto , Exones/genética , Femenino , Crecimiento/fisiología , Homocigoto , Humanos , Péptidos y Proteínas de Señalización Intracelular , Enfermedades Renales/patología , Mutación/genética , Análisis de Secuencia de ADN
4.
Hum Mutat ; 34(5): 714-24, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23418020

RESUMEN

Ciliopathies are genetically heterogeneous disorders characterized by variable expressivity and overlaps between different disease entities. This is exemplified by the short rib-polydactyly syndromes, Jeune, Sensenbrenner, and Mainzer-Saldino chondrodysplasia syndromes. These three syndromes are frequently caused by mutations in intraflagellar transport (IFT) genes affecting the primary cilia, which play a crucial role in skeletal and chondral development. Here, we identified mutations in IFT140, an IFT complex A gene, in five Jeune asphyxiating thoracic dystrophy (JATD) and two Mainzer-Saldino syndrome (MSS) families, by screening a cohort of 66 JATD/MSS patients using whole exome sequencing and targeted resequencing of a customized ciliopathy gene panel. We also found an enrichment of rare IFT140 alleles in JATD compared with nonciliopathy diseases, implying putative modifier effects for certain alleles. IFT140 patients presented with mild chest narrowing, but all had end-stage renal failure under 13 years of age and retinal dystrophy when examined for ocular dysfunction. This is consistent with the severe cystic phenotype of Ift140 conditional knockout mice, and the higher level of Ift140 expression in kidney and retina compared with the skeleton at E15.5 in the mouse. IFT140 is therefore a major cause of cono-renal syndromes (JATD and MSS). The present study strengthens the rationale for IFT140 screening in skeletal ciliopathy spectrum patients that have kidney disease and/or retinal dystrophy.


Asunto(s)
Transporte Biológico/genética , Cilios/metabolismo , Enfermedades Renales/genética , Mutación , Animales , Ataxia Cerebelosa/genética , Niño , Estudios de Cohortes , Progresión de la Enfermedad , Exoma , Humanos , Enfermedades Renales/patología , Masculino , Ratones , Retinitis Pigmentosa/genética
5.
J Am Soc Nephrol ; 22(11): 2047-56, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22034641

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is typically a late-onset disease caused by mutations in PKD1 or PKD2, but about 2% of patients with ADPKD show an early and severe phenotype that can be clinically indistinguishable from autosomal recessive polycystic kidney disease (ARPKD). The high recurrence risk in pedigrees with early and severe PKD strongly suggests a common familial modifying background, but the mechanisms underlying the extensive phenotypic variability observed among affected family members remain unknown. Here, we describe severely affected patients with PKD who carry, in addition to their expected familial germ-line defect, additional mutations in PKD genes, including HNF-1ß, which likely aggravate the phenotype. Our findings are consistent with a common pathogenesis and dosage theory for PKD and may propose a general concept for the modification of disease expression in other so-called monogenic disorders.


Asunto(s)
Translocador Nuclear del Receptor de Aril Hidrocarburo/genética , Riñón Poliquístico Autosómico Dominante/genética , Índice de Severidad de la Enfermedad , Canales Catiónicos TRPP/genética , Adulto , Edad de Inicio , Anciano , Secuencia de Aminoácidos , Salud de la Familia , Femenino , Genotipo , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Fenotipo , Riñón Poliquístico Autosómico Dominante/epidemiología , Riñón Poliquístico Autosómico Dominante/patología , Embarazo , Factores de Riesgo
6.
Am J Hum Genet ; 82(4): 959-70, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18371931

RESUMEN

Many genetic diseases have been linked to the dysfunction of primary cilia, which occur nearly ubiquitously in the body and act as solitary cellular mechanosensory organelles. The list of clinical manifestations and affected tissues in cilia-related disorders (ciliopathies) such as nephronophthisis is broad and has been attributed to the wide expression pattern of ciliary proteins. However, little is known about the molecular mechanisms leading to this dramatic diversity of phenotypes. We recently reported hypomorphic NPHP3 mutations in children and young adults with isolated nephronophthisis and associated hepatic fibrosis or tapetoretinal degeneration. Here, we chose a combinatorial approach in mice and humans to define the phenotypic spectrum of NPHP3/Nphp3 mutations and the role of the nephrocystin-3 protein. We demonstrate that the pcy mutation generates a hypomorphic Nphp3 allele that is responsible for the cystic kidney disease phenotype, whereas complete loss of Nphp3 function results in situs inversus, congenital heart defects, and embryonic lethality in mice. In humans, we show that NPHP3 mutations can cause a broad clinical spectrum of early embryonic patterning defects comprising situs inversus, polydactyly, central nervous system malformations, structural heart defects, preauricular fistulas, and a wide range of congenital anomalies of the kidney and urinary tract (CAKUT). On the functional level, we show that nephrocystin-3 directly interacts with inversin and can inhibit like inversin canonical Wnt signaling, whereas nephrocystin-3 deficiency leads in Xenopus laevis to typical planar cell polarity defects, suggesting a role in the control of canonical and noncanonical (planar cell polarity) Wnt signaling.


Asunto(s)
Anomalías Múltiples/genética , Muerte Fetal/genética , Enfermedades Renales Quísticas/genética , Cinesinas/genética , Situs Inversus/genética , Adolescente , Animales , Niño , Femenino , Humanos , Recién Nacido , Riñón/anomalías , Cinesinas/metabolismo , Hígado/anomalías , Masculino , Ratones , Ratones Mutantes , Mutación , Páncreas/anomalías , Linaje , Síndrome , Factores de Transcripción/metabolismo , Proteínas Wnt/metabolismo , Xenopus laevis
7.
Hum Mutat ; 29(1): 45-52, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17705300

RESUMEN

Meckel-Gruber syndrome (MKS) is an autosomal recessive, lethal multisystemic disorder characterized by meningooccipital encephalocele, cystic kidney dysplasia, hepatobiliary ductal plate malformation, and postaxial polydactyly. Recently, genes for MKS1 and MKS3 were identified, putting MKS on the list of ciliary disorders (ciliopathies). By positional cloning in a distantly related multiplex family, we mapped a novel locus for MKS to a 3-Mb interval on 12q21. Sequencing of the CEP290 gene located in the minimal critical region showed a homozygous 1-bp deletion supposed to lead to loss of function of the encoded centrosomal protein CEP290/nephrocystin-6. CEP290 is thought to be involved in chromosome segregation and localizes to cilia, centrosomes, and the nucleus. Subsequent analysis of another consanguineous multiplex family revealed homozygous haplotypes and the same frameshift mutation. Our findings add to the increasing body of evidence that ciliopathies can cause a broad spectrum of disease phenotypes, and pleiotropic effects of CEP290 mutations range from single organ involvement with isolated Leber congenital amaurosis to Joubert syndrome and lethal early embryonic multisystemic malformations in Meckel-Gruber syndrome. We compiled clinical and genetic data of all patients with CEP290 mutations described so far. No clear-cut genotype-phenotype correlations were apparent as almost all mutations are nonsense, frameshift, or splice-site changes and scattered throughout the gene irrespective of the patients' phenotypes. Conclusively, other factors than the type and location of CEP290 mutations may underlie phenotypic variability.


Asunto(s)
Anomalías Múltiples/genética , Antígenos de Neoplasias/genética , Mutación , Proteínas de Neoplasias/genética , Secuencia de Aminoácidos , Secuencia de Bases , Proteínas de Ciclo Celular , Sistema Nervioso Central/anomalías , Proteínas del Citoesqueleto , Análisis Mutacional de ADN , Haplotipos , Humanos , Enfermedades Renales Quísticas/genética , Hígado/anomalías , Datos de Secuencia Molecular , Linaje , Polimorfismo de Nucleótido Simple , Síndrome
8.
Hum Mutat ; 28(6): 638-9, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17437276

RESUMEN

Meckel-Gruber syndrome (MKS) is an autosomal recessive, usually lethal multisystemic disorder characterized by early developmental anomalies of the central nervous system, cystic kidney dysplasia, hepatobiliary ductal plate malformation, and postaxial polydactyly. Three MKS loci have been mapped and recently, two genes were identified: MKS1 on 17q22 in Caucasian kindreds and MKS3 on 8q22 in Omani and Pakistani families, putting MKS on the growing list of ciliary disorders ("ciliopathies"). We performed linkage analysis for MKS1-3 in 14 consanguineous and/or multiplex families of different ethnic origins with histologic diagnosis and at least three classic MKS manifestations in each kindred. Unexpectedly, only five families were linked to any of the known MKS loci, clearly indicating further locus heterogeneity. All five families showed homozygosity for MKS1 and, intriguingly, were of non-Caucasian origin. MKS1 sequencing revealed no mutation in two of these pedigrees, whereas different, novel splicing defects were identified in the three other families and an additional sporadic German patient. Given that all of our mutations and two of the in total four known MKS1 changes cause aberrant splicing (while the other two known mutations were frameshift mutations), we hypothesize that splicing defects are a crucial mutational mechanism in MKS1 which apparently is one of the main loci and key players in MKS. Our results indicate that MKS1 mutations are not restricted to the Caucasian gene pool and suggest further genetic heterogeneity for MKS. Overall, our data have immediate implications for genetic counselling and testing approaches in MKS.


Asunto(s)
Anomalías Múltiples/genética , Empalme Alternativo , Sistema Nervioso Central/anomalías , Enfermedades Renales Quísticas/genética , Mutación , Proteínas/genética , Consanguinidad , Análisis Mutacional de ADN , Exones , Haplotipos , Homocigoto , Humanos , Intrones , Kuwait , Hígado/anomalías , Linaje , Polidactilia/genética , Eliminación de Secuencia , Síndrome , Turquía , Población Blanca/genética
10.
Nat Genet ; 49(7): 1025-1034, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28530676

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD), usually considered to be a genetically homogeneous disease caused by mutations in PKHD1, has been associated with ciliary dysfunction. Here, we describe mutations in DZIP1L, which encodes DAZ interacting protein 1-like, in patients with ARPKD. We further validated these findings through loss-of-function studies in mice and zebrafish. DZIP1L localizes to centrioles and to the distal ends of basal bodies, and interacts with septin2, a protein implicated in maintenance of the periciliary diffusion barrier at the ciliary transition zone. In agreement with a defect in the diffusion barrier, we found that the ciliary-membrane translocation of the PKD proteins polycystin-1 and polycystin-2 is compromised in DZIP1L-mutant cells. Together, these data provide what is, to our knowledge, the first conclusive evidence that ARPKD is not a homogeneous disorder and further establish DZIP1L as a second gene involved in ARPKD pathogenesis.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo/genética , Anomalías Múltiples/embriología , Anomalías Múltiples/genética , Proteínas Adaptadoras Transductoras de Señales/deficiencia , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Centriolos/metabolismo , Cromosomas Humanos Par 3/genética , Cilios/metabolismo , Consanguinidad , Modelos Animales de Enfermedad , Embrión no Mamífero/anomalías , Femenino , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Humanos , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Linaje , Riñón Poliquístico Autosómico Recesivo/embriología , Transporte de Proteínas , Septinas/metabolismo , Canales Catiónicos TRPP/metabolismo , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología
12.
PLoS One ; 10(2): e0116680, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25646624

RESUMEN

Renal cysts are clinically and genetically heterogeneous conditions. Autosomal dominant polycystic kidney disease (ADPKD) is the most frequent life-threatening genetic disease and mainly caused by mutations in PKD1. The presence of six PKD1 pseudogenes and tremendous allelic heterogeneity make molecular genetic testing challenging requiring laborious locus-specific amplification. Increasing evidence suggests a major role for PKD1 in early and severe cases of ADPKD and some patients with a recessive form. Furthermore it is becoming obvious that clinical manifestations can be mimicked by mutations in a number of other genes with the necessity for broader genetic testing. We established and validated a sequence capture based NGS testing approach for all genes known for cystic and polycystic kidney disease including PKD1. Thereby, we demonstrate that the applied standard mapping algorithm specifically aligns reads to the PKD1 locus and overcomes the complication of unspecific capture of pseudogenes. Employing careful and experienced assessment of NGS data, the method is shown to be very specific and equally sensitive as established methods. An additional advantage over conventional Sanger sequencing is the detection of copy number variations (CNVs). Sophisticated bioinformatic read simulation increased the high analytical depth of the validation study and further demonstrated the strength of the approach. We further raise some awareness of limitations and pitfalls of common NGS workflows when applied in complex regions like PKD1 demonstrating that quality of NGS needs more than high coverage of the target region. By this, we propose a time- and cost-efficient diagnostic strategy for comprehensive molecular genetic testing of polycystic kidney disease which is highly automatable and will be of particular value when therapeutic options for PKD emerge and genetic testing is needed for larger numbers of patients.


Asunto(s)
Técnicas de Diagnóstico Molecular/métodos , Enfermedades Renales Poliquísticas/diagnóstico , Enfermedades Renales Poliquísticas/genética , Variaciones en el Número de Copia de ADN , Reacciones Falso Negativas , Duplicación de Gen , Sitios Genéticos/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Riñón/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Seudogenes/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora
13.
Clin J Am Soc Nephrol ; 9(10): 1729-36, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25104275

RESUMEN

BACKGROUND AND OBJECTIVES: Autosomal recessive polycystic kidney disease (ARPKD) is caused by mutations in the PKHD1 gene. The longest open reading frame comprises 66 exons encoding polyductin or fibrocystin, a type I transmembrane protein with 4074 amino acids. Functional investigations are considerably hampered by its large size and lack of expression in tissues that are usually available for analysis such as lymphocytes or fibroblasts. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: Allegedly strong and clear-cut genotype-phenotype correlations for the type of PKHD1 mutation could be established. Thus far, practically all patients with two truncating mutations showed perinatal or neonatal demise and at least one hypomorphic missense mutation is thought to be indispensable for survival. Mutation analysis of >500 ARPKD families was performed by conventional and next-generation sequencing techniques. RESULTS: This study presents four unrelated patients with ARPKD with a nonlethal, moderate clinical course despite the burden of two PKHD1 mutations expected to lead to premature termination of translation. In line with parental consanguinity, all mutations occurred in the homozygous state and segregated with the disorder in these families. To try to unravel the mechanisms that underlie this obvious contradiction, these patients were further analyzed in detail by utilizing different methods. In all cases, complex transcriptional alterations were detected. Alternative splicing patterns might disrupt a critical stoichiometric and temporal balance between different protein products and may play a crucial role in defining the phenotype of these patients. CONCLUSIONS: Although these findings represent rare events, they are of importance for genetic counseling and illustrate that some caution is warranted in the interpretation of mutations and their clinical significance. The authors hypothesize that expression of a minimal amount of functional protein is needed for survival of the neonatal period in ARPKD.


Asunto(s)
Mutación , Riñón Poliquístico Autosómico Recesivo/genética , Receptores de Superficie Celular/genética , Transcripción Genética , Animales , Células COS , Niño , Preescolar , Chlorocebus aethiops , Biología Computacional , Consanguinidad , Análisis Mutacional de ADN , Progresión de la Enfermedad , Exones , Femenino , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Masculino , Linaje , Fenotipo , Riñón Poliquístico Autosómico Recesivo/diagnóstico , Riñón Poliquístico Autosómico Recesivo/metabolismo , Riñón Poliquístico Autosómico Recesivo/terapia , Pronóstico , Receptores de Superficie Celular/metabolismo , Transfección
14.
Nat Genet ; 45(8): 951-6, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23793029

RESUMEN

Nephronophthisis is an autosomal recessive cystic kidney disease that leads to renal failure in childhood or adolescence. Most NPHP gene products form molecular networks. Here we identify ANKS6 as a new NPHP family member that connects NEK8 (NPHP9) to INVS (NPHP2) and NPHP3. We show that ANKS6 localizes to the proximal cilium and confirm its role in renal development through knockdown experiments in zebrafish and Xenopus laevis. We also identify six families with ANKS6 mutations affected by nephronophthisis, including severe cardiovascular abnormalities, liver fibrosis and situs inversus. The oxygen sensor HIF1AN hydroxylates ANKS6 and INVS and alters the composition of the ANKS6-INVS-NPHP3 module. Knockdown of Hif1an in Xenopus results in a phenotype that resembles loss of other NPHP proteins. Network analyses uncovered additional putative NPHP proteins and placed ANKS6 at the center of this NPHP module, explaining the overlapping disease manifestation caused by mutation in ANKS6, NEK8, INVS or NPHP3.


Asunto(s)
Enfermedades Renales Quísticas/genética , Cinesinas/genética , Proteínas Nucleares/genética , Proteínas Quinasas/genética , Factores de Transcripción/genética , Animales , Cilios/metabolismo , Consanguinidad , Exones , Técnicas de Silenciamiento del Gen , Humanos , Intrones , Enfermedades Renales Quísticas/metabolismo , Cinesinas/metabolismo , Ratones , Mutación , Quinasas Relacionadas con NIMA , Proteínas Nucleares/metabolismo , Fenotipo , Enfermedades Renales Poliquísticas/genética , Unión Proteica , Mapas de Interacción de Proteínas , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Factores de Transcripción/metabolismo , Xenopus/embriología , Xenopus/metabolismo , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
16.
J Invest Dermatol ; 128(4): 791-6, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17914448

RESUMEN

Congenital anonychia is a rare autosomal-recessive disorder characterized by the absence of finger- and toenails. Recently, we and others identified the secreted Wnt signaling ligand R-spondin 4 (RSPO4) as the first gene known to be responsible for inherited anonychia. R-spondins are secreted proteins that activate the Wnt/beta-catenin signaling pathway. This puts anonychia on the growing list of congenital malformation syndromes caused by Wnt signaling pathway defects. Here, we expand the RSPO4 mutational spectrum by identification of the previously unknown mutations c.190C>T (p.Arg64Cys) in exon 2 and c.301C>T (p.Gln101X) in exon 3, thereby corroborating R-spondin 4 as the major protein in autosomal-recessive anonychia. Almost all RSPO4 mutations detected so far affect the highly conserved exons 2 and 3. Thus, we postulate that RSPO4 mutations preferentially cluster in the furin-like cysteine-rich domains of R-spondin 4, which is in line with experimental data proposing that for beta-catenin stabilization, a shortened protein comprising just these two regions is sufficient.


Asunto(s)
Uñas Malformadas/genética , Trombospondinas/genética , Proteínas Wnt/metabolismo , Adolescente , Secuencia de Aminoácidos , Preescolar , Cisteína/química , Femenino , Furina/química , Ligamiento Genético , Humanos , Ligandos , Masculino , Datos de Secuencia Molecular , Mutación , Linaje , Estructura Terciaria de Proteína , Trombospondinas/química , Trombospondinas/metabolismo , beta Catenina/metabolismo
18.
J Hum Genet ; 51(9): 788-793, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16897190

RESUMEN

Autosomal recessive polycystic kidney disease (ARPKD) is caused by mutations in the PKHD1 (polycystic kidney and hepatic disease 1) gene on chromosome 6p12. The longest continuous open reading frame comprises 66 exons encoding a novel 4,074 aa multidomain integral membrane protein (polyductin/fibrocystin) of unknown function. Various alternatively spliced transcripts may additionally result in different isoproteins. Overall, the large size of PKHD1, its complex pattern of splicing, multiple allelism and lack of knowledge of the encoded protein's/proteins' functions pose significant challenges to DNA-based diagnostic testing. Nucleotide substitutions, particularly if residing in regulatory elements or introns outside the splice consensus sites, are often difficult to assess without further functional analyses and cannot be unambiguously classified as disease-associated. Investigations on the transcript level, however, are hampered as PKHD1 is not widely expressed in blood lymphocytes. We thus determined the functional significance of the novel splice site mutation c.53-3C>A in intron 2 by RNA analyses by minigene-construction. The mutant allele was shown to cause skipping of exon 3. Thus, given the minigene results together with 400 control chromosomes negative for this change, segregation of the mutation with the phenotype, and a significant lowering of the strength of the splice site by bioinformatics, the mutant allele is most likely pathogenic. To the best of our knowledge, this is the first study that defines the consequences of a PKHD1 splice mutation and underlines the relevance of functional analyses in determining the pathogenicity of changes of unknown significance.


Asunto(s)
Riñón Poliquístico Autosómico Recesivo/genética , Receptores de Superficie Celular/genética , Secuencia de Bases , ADN Complementario/genética , Femenino , Humanos , Recién Nacido , Datos de Secuencia Molecular , Mutación , Sitios de Empalme de ARN
19.
Mol Diagn Ther ; 10(3): 163-74, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16771602

RESUMEN

Cystic kidney diseases (CKDs) are a clinically and genetically heterogeneous group of disorders characterized by progressive fibrocystic renal and hepatobiliary changes. Recent findings have proven the cystogenic process to be compatible with cellular dedifferentiation, i. e. increased apoptosis and proliferation rates, altered protein sorting and secretory characteristics, as well as disorganization of the extracellular matrix. Compelling evidence suggests that cilia play a central pathogenic role and most cystic kidney disorders converge into a common pathogenic pathway. Recently, several promising trials have further extended our understanding of the pathophysiology of CKD and may have the potential for rational personalized therapies in future years. This review aims to summarize the current state of knowledge of the structure and function of proteins underlying polycystic kidney disease, to explore the clinical consequences of changes in respective genes, and to discuss potential therapeutic approaches.


Asunto(s)
Enfermedades Renales Quísticas/genética , Genotipo , Humanos , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/tratamiento farmacológico , Enfermedades Renales Quísticas/fisiopatología , Fenotipo , Riñón Poliquístico Autosómico Dominante/diagnóstico , Riñón Poliquístico Autosómico Dominante/tratamiento farmacológico , Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/fisiopatología , Riñón Poliquístico Autosómico Recesivo/diagnóstico , Riñón Poliquístico Autosómico Recesivo/tratamiento farmacológico , Riñón Poliquístico Autosómico Recesivo/genética , Riñón Poliquístico Autosómico Recesivo/fisiopatología , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Canales Catiónicos TRPP/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA