Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Am Soc Nephrol ; 34(6): 1105-1119, 2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-36995132

RESUMEN

SIGNIFICANCE STATEMENT: Congenital obstructive uropathy (COU) is a prevalent human developmental defect with highly heterogeneous clinical presentations and outcomes. Genetics may refine diagnosis, prognosis, and treatment, but the genomic architecture of COU is largely unknown. Comprehensive genomic screening study of 733 cases with three distinct COU subphenotypes revealed disease etiology in 10.0% of them. We detected no significant differences in the overall diagnostic yield among COU subphenotypes, with characteristic variable expressivity of several mutant genes. Our findings therefore may legitimize a genetic first diagnostic approach for COU, especially when burdening clinical and imaging characterization is not complete or available. BACKGROUND: Congenital obstructive uropathy (COU) is a common cause of developmental defects of the urinary tract, with heterogeneous clinical presentation and outcome. Genetic analysis has the potential to elucidate the underlying diagnosis and help risk stratification. METHODS: We performed a comprehensive genomic screen of 733 independent COU cases, which consisted of individuals with ureteropelvic junction obstruction ( n =321), ureterovesical junction obstruction/congenital megaureter ( n =178), and COU not otherwise specified (COU-NOS; n =234). RESULTS: We identified pathogenic single nucleotide variants (SNVs) in 53 (7.2%) cases and genomic disorders (GDs) in 23 (3.1%) cases. We detected no significant differences in the overall diagnostic yield between COU sub-phenotypes, and pathogenic SNVs in several genes were associated to any of the three categories. Hence, although COU may appear phenotypically heterogeneous, COU phenotypes are likely to share common molecular bases. On the other hand, mutations in TNXB were more often identified in COU-NOS cases, demonstrating the diagnostic challenge in discriminating COU from hydronephrosis secondary to vesicoureteral reflux, particularly when diagnostic imaging is incomplete. Pathogenic SNVs in only six genes were found in more than one individual, supporting high genetic heterogeneity. Finally, convergence between data on SNVs and GDs suggest MYH11 as a dosage-sensitive gene possibly correlating with severity of COU. CONCLUSIONS: We established a genomic diagnosis in 10.0% of COU individuals. The findings underscore the urgent need to identify novel genetic susceptibility factors to COU to better define the natural history of the remaining 90% of cases without a molecular diagnosis.


Asunto(s)
Hidronefrosis , Obstrucción Ureteral , Reflujo Vesicoureteral , Humanos , Variaciones en el Número de Copia de ADN , Obstrucción Ureteral/complicaciones , Obstrucción Ureteral/genética , Reflujo Vesicoureteral/diagnóstico , Reflujo Vesicoureteral/genética , Pelvis Renal/patología
2.
Hum Genet ; 142(1): 73-88, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36066768

RESUMEN

Most patients with congenital anomalies of the kidney and urinary tract (CAKUT) remain genetically unexplained. In search of novel genes associated with CAKUT in humans, we applied whole-exome sequencing in a patient with kidney, anorectal, spinal, and brain anomalies, and identified a rare heterozygous missense variant in the DACT1 (dishevelled binding antagonist of beta catenin 1) gene encoding a cytoplasmic WNT signaling mediator. Our patient's features overlapped Townes-Brocks syndrome 2 (TBS2) previously described in a family carrying a DACT1 nonsense variant as well as those of Dact1-deficient mice. Therefore, we assessed the role of DACT1 in CAKUT pathogenesis. Taken together, very rare (minor allele frequency ≤ 0.0005) non-silent DACT1 variants were detected in eight of 209 (3.8%) CAKUT families, significantly more frequently than in controls (1.7%). All seven different DACT1 missense variants, predominantly likely pathogenic and exclusively maternally inherited, were located in the interaction region with DVL2 (dishevelled segment polarity protein 2), and biochemical characterization revealed reduced binding of mutant DACT1 to DVL2. Patients carrying DACT1 variants presented with kidney agenesis, duplex or (multi)cystic (hypo)dysplastic kidneys with hydronephrosis and TBS2 features. During murine development, Dact1 was expressed in organs affected by anomalies in patients with DACT1 variants, including the kidney, anal canal, vertebrae, and brain. In a branching morphogenesis assay, tubule formation was impaired in CRISPR/Cas9-induced Dact1-/- murine inner medullary collecting duct cells. In summary, we provide evidence that heterozygous hypomorphic DACT1 variants cause CAKUT and other features of TBS2, including anomalies of the skeleton, brain, distal digestive and genital tract.


Asunto(s)
Sistema Urinario , Anomalías Urogenitales , Humanos , Ratones , Animales , Anomalías Urogenitales/genética , Riñón/anomalías , Sistema Urinario/anomalías , Proteínas Nucleares/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Dishevelled/genética
3.
Genet Med ; 24(2): 384-397, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34906446

RESUMEN

PURPOSE: We aimed to investigate the molecular basis underlying a novel phenotype including hypopituitarism associated with primary ovarian insufficiency. METHODS: We used next-generation sequencing to identify variants in all pedigrees. Expression of Rnpc3/RNPC3 was analyzed by in situ hybridization on murine/human embryonic sections. CRISPR/Cas9 was used to generate mice carrying the p.Leu483Phe pathogenic variant in the conserved murine Rnpc3 RRM2 domain. RESULTS: We described 15 patients from 9 pedigrees with biallelic pathogenic variants in RNPC3, encoding a specific protein component of the minor spliceosome, which is associated with a hypopituitary phenotype, including severe growth hormone (GH) deficiency, hypoprolactinemia, variable thyrotropin (also known as thyroid-stimulating hormone) deficiency, and anterior pituitary hypoplasia. Primary ovarian insufficiency was diagnosed in 8 of 9 affected females, whereas males had normal gonadal function. In addition, 2 affected males displayed normal growth when off GH treatment despite severe biochemical GH deficiency. In both mouse and human embryos, Rnpc3/RNPC3 was expressed in the developing forebrain, including the hypothalamus and Rathke's pouch. Female Rnpc3 mutant mice displayed a reduction in pituitary GH content but with no reproductive impairment in young mice. Male mice exhibited no obvious phenotype. CONCLUSION: Our findings suggest novel insights into the role of RNPC3 in female-specific gonadal function and emphasize a critical role for the minor spliceosome in pituitary and ovarian development and function.


Asunto(s)
Hipopituitarismo , Insuficiencia Ovárica Primaria , Animales , Femenino , Humanos , Hipopituitarismo/genética , Masculino , Ratones , Proteínas Nucleares/genética , Linaje , Fenotipo , Insuficiencia Ovárica Primaria/genética , Prolactina/genética , Proteínas de Unión al ARN/genética
4.
Am J Kidney Dis ; 76(4): 460-470, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32359821

RESUMEN

RATIONALE & OBJECTIVE: Hereditary nephropathies are clinically and genetically heterogeneous disorders. For some patients, the clinical phenotype corresponds to a specific hereditary disease but genetic testing reveals that the expected genotype is not present (phenocopy). The aim of this study was to evaluate the spectrum and frequency of phenocopies identified by using exome sequencing in a cohort of patients who were clinically suspected to have hereditary kidney disorders. STUDY DESIGN: Cross-sectional cohort study. SETTING & PARTICIPANTS: 174 unrelated patients were recruited for exome sequencing and categorized into 7 disease groups according to their clinical presentation. They included autosomal dominant tubulointerstitial kidney disease, Alport syndrome, congenital anomalies of the kidney and urinary tract, ciliopathy, focal segmental glomerulosclerosis/steroid-resistant nephrotic syndrome, VACTERL association, and "other." RESULTS: A genetic diagnosis (either likely pathogenic or pathogenic variant according to the guidelines of the American College of Medical Genetics) was established using exome sequencing in 52 of 174 (30%) cases. A phenocopy was identified for 10 of the 52 exome sequencing-solved cases (19%), representing 6% of the total cohort. The most frequent phenocopies (n=5) were associated with genetic Alport syndrome presenting clinically as focal segmental glomerulosclerosis/steroid-resistant nephrotic syndrome. Strictly targeted gene panels (<25 kilobases) did not identify any of the phenocopy cases. LIMITATIONS: The spectrum of described phenocopies is small. Selection bias may have altered the diagnostic yield within disease groups in our study population. The study cohort was predominantly of non-Finnish European descent, limiting generalizability. Certain hereditary kidney diseases cannot be diagnosed by using exome sequencing (eg, MUC1-autosomal dominant tubulointerstitial kidney disease). CONCLUSIONS: Phenocopies led to the recategorization of disease and altered clinical management. This study highlights that exome sequencing can detect otherwise occult genetic heterogeneity of kidney diseases.


Asunto(s)
Secuenciación del Exoma , Enfermedades Renales/genética , Fenotipo , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Estudios Transversales , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Adulto Joven
5.
Am J Hum Genet ; 98(6): 1228-1234, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27210743

RESUMEN

Nephrolithiasis, a condition in which urinary supersaturation leads to stone formation in the urinary system, affects about 5%-10% of individuals worldwide at some point in their lifetime and results in significant medical costs and morbidity. To date, mutations in more than 30 genes have been described as being associated with nephrolithiasis, and these mutations explain about 15% of kidney stone cases, suggesting that additional nephrolithiasis-associated genes remain to be discovered. To identify additional genes whose mutations are linked to nephrolithiasis, we performed targeted next-generation sequencing of 18 hypothesized candidate genes in 348 unrelated individuals with kidney stones. We detected biallelic mutations in SLC26A1 (solute carrier family 26 member 1) in two unrelated individuals with calcium oxalate kidney stones. We show by immunofluorescence, immunoblotting, and glycosylation analysis that the variant protein mimicking p.Thr185Met has defects in protein folding or trafficking. In addition, by measuring anion exchange activity of SLC26A1, we demonstrate that all the identified mutations in SLC26A1 result in decreased transporter activity. Our data identify SLC26A1 mutations as causing a recessive Mendelian form of nephrolithiasis.


Asunto(s)
Proteínas de Transporte de Anión/genética , Proteínas de Transporte de Anión/metabolismo , Mutación/genética , Nefrolitiasis/etiología , Secuencia de Aminoácidos , Proteínas de Transporte de Anión/química , Bicarbonatos/metabolismo , Técnica del Anticuerpo Fluorescente , Glicosilación , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Immunoblotting , Nefrolitiasis/patología , Conformación Proteica , Pliegue de Proteína , Transporte de Proteínas , Reacción en Cadena en Tiempo Real de la Polimerasa , Homología de Secuencia de Aminoácido , Transportadores de Sulfato , Sulfatos/metabolismo
6.
Am J Med Genet A ; 179(7): 1310-1314, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31012264

RESUMEN

A 16-year-old girl with a history of nontraumatic swelling of both forearms, osteochondromas of the knees, heterotopic ossification of the neck and back, severe malformations of all digits with hypoplastic or absent nails, alopecia partialis of the scalp, and moderate cognitive impairment was seen for diagnostic evaluation. Whole exome sequencing identified an activating mutation of ACVR1 (c.983G > A; p.Gly328Glu) which confirmed a suspected FOP variant. The delayed diagnosis of an FOP variant in this patient could have been avoided if the significance of severe digital malformations had been recognized, especially in the setting of progressive heterotopic ossification.


Asunto(s)
Dedos/anomalías , Mutación , Miositis Osificante/patología , Receptores de Activinas Tipo I/genética , Adolescente , Femenino , Humanos , Miositis Osificante/genética
7.
Kidney Int ; 93(1): 204-213, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28893421

RESUMEN

The incidence of nephrolithiasis continues to rise. Previously, we showed that a monogenic cause could be detected in 11.4% of individuals with adult-onset nephrolithiasis or nephrocalcinosis and in 16.7-20.8% of individuals with onset before 18 years of age, using gene panel sequencing of 30 genes known to cause nephrolithiasis/nephrocalcinosis. To overcome the limitations of panel sequencing, we utilized whole exome sequencing in 51 families, who presented before age 25 years with at least one renal stone or with a renal ultrasound finding of nephrocalcinosis to identify the underlying molecular genetic cause of disease. In 15 of 51 families, we detected a monogenic causative mutation by whole exome sequencing. A mutation in seven recessive genes (AGXT, ATP6V1B1, CLDN16, CLDN19, GRHPR, SLC3A1, SLC12A1), in one dominant gene (SLC9A3R1), and in one gene (SLC34A1) with both recessive and dominant inheritance was detected. Seven of the 19 different mutations were not previously described as disease-causing. In one family, a causative mutation in one of 117 genes that may represent phenocopies of nephrolithiasis-causing genes was detected. In nine of 15 families, the genetic diagnosis may have specific implications for stone management and prevention. Several factors that correlated with the higher detection rate in our cohort were younger age at onset of nephrolithiasis/nephrocalcinosis, presence of multiple affected members in a family, and presence of consanguinity. Thus, we established whole exome sequencing as an efficient approach toward a molecular genetic diagnosis in individuals with nephrolithiasis/nephrocalcinosis who manifest before age 25 years.


Asunto(s)
Secuenciación del Exoma , Mutación , Nefrocalcinosis/genética , Nefrolitiasis/genética , Adolescente , Edad de Inicio , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Estudios de Asociación Genética , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Herencia , Humanos , Lactante , Masculino , Nefrocalcinosis/diagnóstico por imagen , Nefrocalcinosis/epidemiología , Nefrolitiasis/diagnóstico por imagen , Nefrolitiasis/epidemiología , Linaje , Fenotipo , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo , Tomografía Computarizada por Rayos X , Ultrasonografía , Adulto Joven
8.
Nephrol Dial Transplant ; 33(1): 85-94, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-27708066

RESUMEN

Background: Lowe syndrome (LS) and Dent-2 disease (DD2) are disorders associated with mutations in the OCRL gene and characterized by progressive chronic kidney disease (CKD). Here, we aimed to investigate the long-term renal outcome and identify potential determinants of CKD and its progression in children with these tubulopathies. Methods: Retrospective analyses were conducted of clinical and genetic data in a cohort of 106 boys (LS: 88 and DD2: 18). For genotype-phenotype analysis, we grouped mutations according to their type and localization. To investigate progression of CKD we used survival analysis by Kaplan-Meier method using stage 3 CKD as the end-point. Results: Median estimated glomerular filtration rate (eGFR) was lower in the LS group compared with DD2 (58.8 versus 87.4 mL/min/1.73 m2, P < 0.01). CKD stage II-V was found in 82% of patients, of these 58% and 28% had moderate-to-severe CKD in LS and DD2, respectively. Three patients (3%), all with LS, developed stage 5 of CKD. Survival analysis showed that LS was also associated with a faster CKD progression than DD2 (P < 0.01). On multivariate analysis, eGFR was dependent only on age (b = -0.46, P < 0.001). Localization, but not type of mutations, tended to correlate with eGFR. There was also no significant association between presence of nephrocalcinosis, hypercalciuria, proteinuria and number of adverse clinical events and CKD. Conclusions: CKD is commonly found in children with OCRL mutations. CKD progression was strongly related to the underlying diagnosis but did not associate with clinical parameters, such as nephrocalcinosis or proteinuria.


Asunto(s)
Hipercalciuria/epidemiología , Mutación , Nefrocalcinosis/epidemiología , Monoéster Fosfórico Hidrolasas/genética , Proteinuria/epidemiología , Insuficiencia Renal Crónica/genética , Adolescente , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Genotipo , Tasa de Filtración Glomerular , Humanos , Hipercalciuria/genética , Masculino , Nefrocalcinosis/genética , Fenotipo , Proteinuria/genética , Insuficiencia Renal Crónica/fisiopatología , Insuficiencia Renal Crónica/terapia , Estudios Retrospectivos , Resultado del Tratamiento
9.
Clin Endocrinol (Oxf) ; 87(4): 350-358, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28656605

RESUMEN

OBJECTIVE: To evaluate the safety, local tolerability, pharmacodynamics and pharmacokinetics of escalating single doses of once-weekly somapacitan, a reversible, albumin-binding GH derivative, vs once-daily GH in children with GH deficiency (GHD). DESIGN: Phase 1, randomized, open-label, active-controlled, dose-escalation trial (NCT01973244). PATIENTS: Thirty-two prepubertal GH-treated children with GHD were sequentially randomized 3:1 within each of four cohorts to a single dose of somapacitan (0.02, 0.04, 0.08 and 0.16 mg/kg; n=6 each), or once-daily Norditropin® SimpleXx® (0.03 mg/kg; n=2 each) for 7 days. MEASUREMENTS: Pharmacokinetic and pharmacodynamic profiles were assessed. RESULTS: Adverse events were all mild, and there were no apparent treatment-dependent patterns in type or frequency. Four mild transient injection site reactions were reported in three of 24 children treated with somapacitan. No antisomapacitan/anti-human growth hormone (hGH) antibodies were detected. Mean serum concentrations of somapacitan increased in a dose-dependent but nonlinear manner: maximum concentration ranged from 21.8 ng/mL (0.02 mg/kg dose) to 458.4 ng/mL (0.16 mg/kg dose). IGF-I and IGFBP-3, and change from baseline in IGF-I standard deviation score (SDS) and IGFBP-3 SDS, increased dose dependently; greatest changes in SDS values were seen for 0.16 mg/kg. IGF-I SDS values were between -2 and +2 SDS, except for peak IGF-I SDS with 0.08 mg/kg somapacitan. Postdosing, IGF-I SDS remained above baseline levels for at least 1 week. CONCLUSIONS: Single doses of once-weekly somapacitan (0.02-0.16 mg/kg) were well tolerated in children with GHD, with IGF-I profiles supporting a once-weekly treatment profile. No clinically significant safety/tolerability signals or immunogenicity concerns were identified.


Asunto(s)
Enanismo Hipofisario/tratamiento farmacológico , Hormona del Crecimiento/uso terapéutico , Hormona de Crecimiento Humana/deficiencia , Niño , Preescolar , Enanismo Hipofisario/sangre , Femenino , Hormona del Crecimiento/análogos & derivados , Hormona del Crecimiento/farmacocinética , Hormona de Crecimiento Humana/análogos & derivados , Hormona de Crecimiento Humana/sangre , Humanos , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino
10.
N Engl J Med ; 369(7): 621-9, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23862974

RESUMEN

BACKGROUND: Congenital abnormalities of the kidney and the urinary tract are the most common cause of pediatric kidney failure. These disorders are highly heterogeneous, and the etiologic factors are poorly understood. METHODS: We performed genomewide linkage analysis and whole-exome sequencing in a family with an autosomal dominant form of congenital abnormalities of the kidney or urinary tract (seven affected family members). We also performed a sequence analysis in 311 unrelated patients, as well as histologic and functional studies. RESULTS: Linkage analysis identified five regions of the genome that were shared among all affected family members. Exome sequencing identified a single, rare, deleterious variant within these linkage intervals, a heterozygous splice-site mutation in the dual serine-threonine and tyrosine protein kinase gene (DSTYK). This variant, which resulted in aberrant splicing of messenger RNA, was present in all affected family members. Additional, independent DSTYK mutations, including nonsense and splice-site mutations, were detected in 7 of 311 unrelated patients. DSTYK is highly expressed in the maturing epithelia of all major organs, localizing to cell membranes. Knockdown in zebrafish resulted in developmental defects in multiple organs, which suggested loss of fibroblast growth factor (FGF) signaling. Consistent with this finding is the observation that DSTYK colocalizes with FGF receptors in the ureteric bud and metanephric mesenchyme. DSTYK knockdown in human embryonic kidney cells inhibited FGF-stimulated phosphorylation of extracellular-signal-regulated kinase (ERK), the principal signal downstream of receptor tyrosine kinases. CONCLUSIONS: We detected independent DSTYK mutations in 2.3% of patients with congenital abnormalities of the kidney or urinary tract, a finding that suggests that DSTYK is a major determinant of human urinary tract development, downstream of FGF signaling. (Funded by the National Institutes of Health and others.).


Asunto(s)
Mutación , Proteína Serina-Treonina Quinasas de Interacción con Receptores/genética , Sistema Urinario/anomalías , Anomalías Urogenitales/genética , Adulto , Animales , Secuencia de Bases , Niño , Exoma , Femenino , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Estudio de Asociación del Genoma Completo , Heterocigoto , Humanos , Lactante , Riñón/anomalías , Masculino , Ratones , Datos de Secuencia Molecular , Linaje , ARN Interferente Pequeño , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Sistema Urinario/crecimiento & desarrollo , Sistema Urinario/metabolismo , Adulto Joven
11.
J Pediatr ; 177S: S127-S137, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27666261

RESUMEN

Macedonia is a multiethnic developing country with a new democratic political system in transition from a former communist country. The country gained independence as former Yugoslav Republic of Macedonia in 1991. Recent health reforms have privatized pediatric primary care and introduced family doctors alongside primary care pediatricians. Increasing privatization of hospitals have left the state-run hospitals short of pediatric specialists and subspecialists as doctors moved to private hospitals for better salaries and working conditions. There is little coordinated action between the Ministry of Health, health insurance fund, and Macedonian pediatricians to overcome the problems that now exist within the pediatric/child health system because of these recent reforms, which were politically driven without consultation with the Macedonian Pediatric Association. These recent decisions will have an adverse effect on the quality of care for children and families, which will likely continue for another 5-10 years.


Asunto(s)
Servicios de Salud del Niño , Reforma de la Atención de Salud , Calidad de la Atención de Salud , Niño , Preescolar , Grecia , Humanos
12.
J Am Soc Nephrol ; 26(3): 543-51, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25296721

RESUMEN

Nephrolithiasis is a prevalent condition with a high morbidity. Although dozens of monogenic causes have been identified, the fraction of single-gene disease has not been well studied. To determine the percentage of cases that can be molecularly explained by mutations in 1 of 30 known kidney stone genes, we conducted a high-throughput mutation analysis in a cohort of consecutively recruited patients from typical kidney stone clinics. The cohort comprised 272 genetically unresolved individuals (106 children and 166 adults) from 268 families with nephrolithiasis (n=256) or isolated nephrocalcinosis (n=16). We detected 50 likely causative mutations in 14 of 30 analyzed genes, leading to a molecular diagnosis in 14.9% (40 of 268) of all cases; 20 of 50 detected mutations were novel (40%). The cystinuria gene SLC7A9 (n=19) was most frequently mutated. The percentage of monogenic cases was notably high in both the adult (11.4%) and pediatric cohorts (20.8%). Recessive causes were more frequent among children, whereas dominant disease occurred more abundantly in adults. Our study provides an in-depth analysis of monogenic causes of kidney stone disease. We suggest that knowledge of the molecular cause of nephrolithiasis and nephrocalcinosis may have practical implications and might facilitate personalized treatment.


Asunto(s)
Nefrocalcinosis/genética , Nefrolitiasis/genética , Adulto , Niño , Estudios de Cohortes , Análisis Mutacional de ADN , Humanos , Mutación Missense
13.
Pediatr Endocrinol Rev ; 13(4): 749-55, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27464418

RESUMEN

LHX4 mutations are rare in combined pituitary hormone deficiency, and even rarer in isolated GHD. We describe a 14 years old boy who was referred for investigation of short stature. Convergent strabismus, nystagmus was present. At the age of 5 years his gait was unstable. A progressive myopathy ensued. Tests of pituitary reserve showed partial IGHD (8.2 ng/ml). Other pituitary hormones were within normal range. Muscle biopsy showed congenital myopathy of undefined etiology. MRI of the brain revealed the empty sella syndrome. Targeted resequencing with a panel containing probe sets for enrichment and analysis of > 4,800 clinically relevant genes, targeting 12Mb of the human genome revealed the c.250C>T (R84C) LHX4 mutation. His father is healthy, with no myopathy or pituitary deficiencies, but has the same LHX4 mutation. This report extends the range of phenotypes associated with LHX4 gene mutations. To the best of our knowledge, we are the first to report on congenital myopathy in an LHX4 gene mutation. Forthwith, we offer a comprehensive review of the patients published so far with their clinical and genetic characteristics.


Asunto(s)
Proteínas con Homeodominio LIM/genética , Enfermedades Musculares/diagnóstico , Enfermedades Musculares/genética , Mutación/genética , Factores de Transcripción/genética , Adolescente , Humanos , Masculino
14.
Med Arch ; 70(2): 148-50, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-27147792

RESUMEN

BACKGROUND: Achondroplasia is a skeletal dysplasia, the most common cause of rhizomelic dwarfism. CASE PRESENTATION: This is a ten year old boy who was first diagnosed prenatally. He had a mutation c1138G>A in the gene FGFR3 in a heterozygotic constellation. His IGF1 and IGFBP3 levels were normal. Two stimulation tests for growth hormone were performed with values within the reference range. His psychomotor development was adequate for his age except for speech difficulty. He started with recombinant hGH (r-hGH) at the age of 3.4 years in a dose of 0.06 mg/kg. His mean Height SDS (HtSDS) was -2.2. RESULTS: The growth increased to 10 cm/year in the first year of therapy (HtSDS -1.1). It decreased during the second year to 4 cm (HtSDS -1.7) and again increased during the third year to 8 cm/year (HtSDS-1.3). In the next years the growth was constant (6.5, 2.3, 3.5 cm / year). He is still growing in the 3(rd) percentile of the growth curve (HtSDS - 1.2) under GH treatment. The body disproportion remained the same. CONCLUSION: The growth response on GH treatment was satisfactory in the first 4 years of treatment, and the boy still continued to grow. The young age at the start of treatment was also of importance. Our other patients with achondroplasia who started treatment older had a poor response to growth hormone.


Asunto(s)
Acondroplasia/tratamiento farmacológico , Estatura/efectos de los fármacos , Hormona de Crecimiento Humana/uso terapéutico , Proteínas Recombinantes/uso terapéutico , Acondroplasia/fisiopatología , Factores de Edad , Estatura/fisiología , Niño , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Terapia de Reemplazo de Hormonas , Humanos , Masculino , Valores de Referencia , Resultado del Tratamiento
15.
Hum Mol Genet ; 22(8): 1625-31, 2013 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-23335591

RESUMEN

In humans, congenital spinal defects occur with an incidence of 0.5-1 per 1000 live births. One of the most severe syndromes with such defects is spondylocostal dysostosis (SCD). Over the past decade, the genetic basis of several forms of autosomal recessive SCD cases has been solved with the identification of four causative genes (DLL3, MESP2, LFNG and HES7). Autosomal dominant forms of SCD have also been reported, but to date no genetic etiology has been described for these. Here, we have used exome capture and next-generation sequencing to identify a stoploss mutation in TBX6 that segregates with disease in two generations of one family. We show that this mutation has a deleterious effect on the transcriptional activation activity of the TBX6 protein, likely due to haploinsufficiency. In mouse, Tbx6 is essential for the patterning of the vertebral precursor tissues, somites; thus, mutation of TBX6 is likely to be causative of SCD in this family. This is the first identification of the genetic cause of an autosomal dominant form of SCD, and also demonstrates the potential of exome sequencing to identify genetic causes of dominant diseases even in small families with few affected individuals.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Múltiples/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Hernia Diafragmática/genética , Hernia Diafragmática/patología , Somitos/metabolismo , Proteínas de Dominio T Box/genética , Anomalías Múltiples/diagnóstico por imagen , Animales , Tipificación del Cuerpo/genética , Modelos Animales de Enfermedad , Genes Dominantes , Cardiopatías Congénitas/diagnóstico por imagen , Hernia Diafragmática/diagnóstico por imagen , Humanos , Ratones , Mutación , Linaje , Radiografía , Análisis de Secuencia de ADN , Somitos/crecimiento & desarrollo , Proteínas de Dominio T Box/metabolismo
16.
Am J Hum Genet ; 91(6): 987-97, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23159250

RESUMEN

We examined the burden of large, rare, copy-number variants (CNVs) in 192 individuals with renal hypodysplasia (RHD) and replicated findings in 330 RHD cases from two independent cohorts. CNV distribution was significantly skewed toward larger gene-disrupting events in RHD cases compared to 4,733 ethnicity-matched controls (p = 4.8 × 10(-11)). This excess was attributable to known and novel (i.e., not present in any database or in the literature) genomic disorders. All together, 55/522 (10.5%) RHD cases harbored 34 distinct known genomic disorders, which were detected in only 0.2% of 13,839 population controls (p = 1.2 × 10(-58)). Another 32 (6.1%) RHD cases harbored large gene-disrupting CNVs that were absent from or extremely rare in the 13,839 population controls, identifying 38 potential novel or rare genomic disorders for this trait. Deletions at the HNF1B locus and the DiGeorge/velocardiofacial locus were most frequent. However, the majority of disorders were detected in a single individual. Genomic disorders were detected in 22.5% of individuals with multiple malformations and 14.5% of individuals with isolated urinary-tract defects; 14 individuals harbored two or more diagnostic or rare CNVs. Strikingly, the majority of the known CNV disorders detected in the RHD cohort have previous associations with developmental delay or neuropsychiatric diseases. Up to 16.6% of individuals with kidney malformations had a molecular diagnosis attributable to a copy-number disorder, suggesting kidney malformations as a sentinel manifestation of pathogenic genomic imbalances. A search for pathogenic CNVs should be considered in this population for the diagnosis of their specific genomic disorders and for the evaluation of the potential for developmental delay.


Asunto(s)
Variaciones en el Número de Copia de ADN , Enfermedades Renales/congénito , Enfermedades Renales/genética , Estudios de Casos y Controles , Aberraciones Cromosómicas , Estudios de Asociación Genética , Genotipo , Humanos , Anotación de Secuencia Molecular
17.
Ren Fail ; 37(9): 1440-3, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26335537

RESUMEN

BACKGROUND: The objective of this study was to assess clinical course and outcome of children with congenital anomalies of the kidney and urinary tract (CAKUT) who had an attack of acute poststreptococcal glomerulonephritis (PSGN). METHOD: Renal status including blood pressure, proteinuria and glomerular filtration rate was retrospectively analyzed in five children with CAKUT and PSGN at the presentation and during the follow up. RESULTS: In the period 2004-2013, 678 patients were diagnosed and recruited in our CAKUT cohort. During this period, 188 patients were hospitalized with the diagnosis of PSGN. A total of five patients had CAKUT and an episode of PSGN (2.6%). Analysis of the follow-up data revealed that three children fully recovered (bilateral vesicoureteral reflux n = 1, ectopic/hypodysplastic kidney n = 1, ureteropelvic junction obstruction n = 1). One child with bilateral hypodysplasia had progressive worsening of the renal function and has been prepared for renal replacement therapy. Another child with single kidney has stable renal function but has significant rising proteinuria, which was not evident on the routine analysis 2 months before the attack of PSGN. CONCLUSION: Poststreptococcal glomerulonephritis in children is generally benign disease with low mortality in acute stage and excellent medium and long-term prognosis. We analyzed our series of PSGN patients and found that 2.6% had anomaly of the urinary tract. The unfavorable outcome was noted in children with single kidney and bilateral renal impairment.


Asunto(s)
Glomerulonefritis/complicaciones , Riñón/anomalías , Infecciones Estreptocócicas/complicaciones , Sistema Urinario/anomalías , Adolescente , Presión Sanguínea , Niño , Preescolar , Femenino , Tasa de Filtración Glomerular , Humanos , Masculino , Pronóstico , Proteinuria , Estudios Retrospectivos
18.
Pediatr Endocrinol Rev ; 13(1): 468-76, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26540764

RESUMEN

Diagnosis and management of pediatric nephrolithiasis/nephrocalcinosis is a very complex and challenging task for every pediatrician. It is based on correct. disease history taking, which may guide to the mode of inheritance (dominant, recessive, x-linked). Ethnicity and consanguinity should also be investigated since they predispose to high prevalence of certain disorders. One should always begin with cheap and available screening tests. Herein we will review clinical, biochemical, metabolic and genetic characteristics of the inherited diseases which lead to nephrolithiasis/nephrocalcinosis, such as: idiopathic hypercalciuria, renal hypophosphatemia, renal tubular acidosis, idiopathic infantile hypercalcemia, Dent disease, familial hypomagnesemia with hypercalciuria and nephrocalcinosis, hypocitraturia, cystinuria, primary hyperoxaluria and renal hypouricemia. Modern genetic techniques such as next generation sequencing enable nowadays diagnosis of rare disease using only a blood sample, trough massive parallel resequencing of many genes. This is very helpful for anuric patients or on dialysis where blood and urine biochemistry are not informative. Genetic testing also replaces invasive liver biopsy or unpleasant acidification tests and enables prenatal or early postnatal diagnosis.


Asunto(s)
Nefrocalcinosis/genética , Nefrocalcinosis/metabolismo , Nefrolitiasis/genética , Nefrolitiasis/metabolismo , Niño , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedades Renales/complicaciones
19.
Am J Med Genet A ; 164A(7): 1713-33, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24782230

RESUMEN

Somatic mutations in the phosphatidylinositol/AKT/mTOR pathway cause segmental overgrowth disorders. Diagnostic descriptors associated with PIK3CA mutations include fibroadipose overgrowth (FAO), Hemihyperplasia multiple Lipomatosis (HHML), Congenital Lipomatous Overgrowth, Vascular malformations, Epidermal nevi, Scoliosis/skeletal and spinal (CLOVES) syndrome, macrodactyly, and the megalencephaly syndrome, Megalencephaly-Capillary malformation (MCAP) syndrome. We set out to refine the understanding of the clinical spectrum and natural history of these phenotypes, and now describe 35 patients with segmental overgrowth and somatic PIK3CA mutations. The phenotypic data show that these previously described disease entities have considerable overlap, and represent a spectrum. While this spectrum overlaps with Proteus syndrome (sporadic, mosaic, and progressive) it can be distinguished by the absence of cerebriform connective tissue nevi and a distinct natural history. Vascular malformations were found in 15/35 (43%) and epidermal nevi in 4/35 (11%) patients, lower than in Proteus syndrome. Unlike Proteus syndrome, 31/35 (89%) patients with PIK3CA mutations had congenital overgrowth, and in 35/35 patients this was asymmetric and disproportionate. Overgrowth was mild with little postnatal progression in most, while in others it was severe and progressive requiring multiple surgeries. Novel findings include: adipose dysregulation present in all patients, unilateral overgrowth that is predominantly left-sided, overgrowth that affects the lower extremities more than the upper extremities and progresses in a distal to proximal pattern, and in the most severely affected patients is associated with marked paucity of adipose tissue in unaffected areas. While the current data are consistent with some genotype-phenotype correlation, this cannot yet be confirmed.


Asunto(s)
Estudios de Asociación Genética , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Tejido Adiposo/patología , Adolescente , Adulto , Edad de Inicio , Niño , Preescolar , Fosfatidilinositol 3-Quinasa Clase I , Femenino , Genotipo , Humanos , Hiperplasia/diagnóstico , Hiperplasia/genética , Lactante , Recién Nacido , Lipoma/diagnóstico , Lipoma/genética , Masculino , Persona de Mediana Edad , Anomalías Musculoesqueléticas/diagnóstico , Anomalías Musculoesqueléticas/genética , Mutación , Nevo/diagnóstico , Nevo/genética , Especificidad de Órganos/genética , Malformaciones Vasculares/diagnóstico , Malformaciones Vasculares/genética , Adulto Joven
20.
Ren Fail ; 36(4): 619-22, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24502542

RESUMEN

INTRODUCTION: The tricho-rhino-phalangeal syndrome type III (TRPS III) is a rare autosomal dominantly inherited condition. The main clinical features are sparse and slow-growing hair and nails, a pear-shaped nose with a bulbous tip, elongated and flat philtrum, thin upper lip, cone-shaped epiphyses of the phalanges, and short stature. All patients have a point mutation in the TRPS1 gene. CASE REPORT: In this paper, we present a 13-year-old female with the typical clinical features of TRPS III, extreme growth retardation, severe deformities of both proximal radii resulting in limited extension of the elbows, and chronic renal failure (CRF) in addition. Molecular diagnostics revealed a missense mutation in exon 6 of TRPS1 that she inherited from her father who is also affected with TRPS III, but does not have CRF. In the index patient, the CRF was found to be due to bilateral renal hypodysplasia (RHD). CONCLUSION: Beside the renal dysplasia, the girl had severe deformities of the proximal radii - findings which have not been reported so far in TRPS III.


Asunto(s)
Dedos/anomalías , Trastornos del Crecimiento/genética , Enfermedades del Cabello/genética , Fallo Renal Crónico/genética , Síndrome de Langer-Giedion/genética , Nariz/anomalías , Anomalías Múltiples/genética , Adolescente , Proteínas de Unión al ADN/genética , Femenino , Humanos , Riñón/anomalías , Mutación Puntual , Radio (Anatomía)/anomalías , Proteínas Represoras , Síndrome , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA