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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 104(1): 35-44, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30554721

RESUMEN

Baratela-Scott syndrome (BSS) is a rare, autosomal-recessive disorder characterized by short stature, facial dysmorphisms, developmental delay, and skeletal dysplasia caused by pathogenic variants in XYLT1. We report clinical and molecular investigation of 10 families (12 individuals) with BSS. Standard sequencing methods identified biallelic pathogenic variants in XYLT1 in only two families. Of the remaining cohort, two probands had no variants and six probands had only a single variant, including four with a heterozygous 3.1 Mb 16p13 deletion encompassing XYLT1 and two with a heterozygous truncating variant. Bisulfite sequencing revealed aberrant hypermethylation in exon 1 of XYLT1, always in trans with the sequence variant or deletion when present; both alleles were methylated in those with no identified variant. Expression of the methylated XYLT1 allele was severely reduced in fibroblasts from two probands. Southern blot studies combined with repeat expansion analysis of genome sequence data showed that the hypermethylation is associated with expansion of a GGC repeat in the XYLT1 promoter region that is not present in the reference genome, confirming that BSS is a trinucleotide repeat expansion disorder. The hypermethylated allele accounts for 50% of disease alleles in our cohort and is not present in 130 control subjects. Our study highlights the importance of investigating non-sequence-based alterations, including epigenetic changes, to identify the missing heritability in genetic disorders.


Asunto(s)
Anomalías Múltiples/genética , Metilación de ADN/genética , Epigénesis Genética/genética , Exones/genética , Mutación , Pentosiltransferasa/genética , Expansión de Repetición de Trinucleótido/genética , Alelos , Southern Blotting , Estudios de Cohortes , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Sulfitos/metabolismo , Síndrome , UDP Xilosa Proteína Xilosiltransferasa
2.
Neurogenetics ; 22(1): 53-64, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33415588

RESUMEN

Proximal spinal muscular atrophy (SMA), a leading genetic cause of infant death worldwide, is an early-onset motor neuron disease characterized by loss of α-motor neurons and associated muscle atrophy. SMA is caused by deletion or other disabling mutations of survival motor neuron 1 (SMN1) but retention of one or more copies of the paralog SMN2. Within the SMA population, there is substantial variation in SMN2 copy number (CN); in general, those individuals with SMA who have a high SMN2 CN have a milder disease. Because SMN2 functions as a disease modifier, its accurate CN determination may have clinical relevance. In this study, we describe the development of array digital PCR (dPCR) to quantify SMN1 and SMN2 CNs in DNA samples using probes that can distinguish the single nucleotide difference between SMN1 and SMN2 in exon 8. This set of dPCR assays can accurately and reliably measure the number of SMN1 and SMN2 copies in DNA samples. In a cohort of SMA patient-derived cell lines, the assay confirmed a strong inverse correlation between SMN2 CN and disease severity. We can detect SMN1-SMN2 gene conversion events in DNA samples by comparing CNs at exon 7 and exon 8. Partial deletions of SMN1 can also be detected with dPCR by comparing CNs at exon 7 or exon 8 with those at intron 1. Array dPCR is a practical technique to determine, accurately and reliably, SMN1 and SMN2 CNs from SMA samples as well as identify gene conversion events and partial deletions of SMN1.


Asunto(s)
Atrofia Muscular Espinal/genética , Mutación/genética , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Conversión Génica/genética , Eliminación de Gen , Humanos , Neuronas Motoras/metabolismo , Fenotipo , Reacción en Cadena de la Polimerasa/métodos , Proteína 2 para la Supervivencia de la Neurona Motora/genética
3.
Pediatr Res ; 90(3): 565-575, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-33446917

RESUMEN

BACKGROUND: Preterm infants with bronchopulmonary dysplasia (BPD) have lifelong increased risk of respiratory morbidities associated with environmental pathogen exposure and underlying mechanisms are poorly understood. The resident immune cells of the lung play vital roles in host defense. However, the effect of perinatal events associated with BPD on pulmonary-specific immune cells is not well understood. METHODS: We used a double-hit model of BPD induced by prenatal chorioamnionitis followed by postnatal hyperoxia, and performed a global transcriptome analysis of all resident pulmonary immune cells. RESULTS: We show significant up-regulation of genes involved in chemokine-mediated signaling and immune cell chemotaxis, and down-regulation of genes involved in multiple T lymphocyte functions. Multiple genes involved in T cell receptor signaling are downregulated and Cd8a gene expression remains downregulated at 2 months of age in spite of recovery in normoxia for 6 weeks. Furthermore, the proportion of CD8a+CD3+ pulmonary immune cells is decreased. CONCLUSIONS: Our study has highlighted that perinatal lung inflammation in a double-hit model of BPD results in short- and long-term dysregulation of genes associated with the pulmonary T cell receptor signaling pathway, which may contribute to increased environmental pathogen-associated respiratory morbidities seen in children and adults with BPD. IMPACT: In a translationally relevant double-hit model of BPD induced by chorioamnionitis and postnatal hyperoxia, we identified pulmonary immune cell-specific transcriptomic changes and showed that T cell receptor signaling genes are downregulated in short term and long term. This is the first comprehensive report delineating transcriptomic changes in resident immune cells of the lung in a translationally relevant double-hit model of BPD. Our study identifies novel resident pulmonary immune cell-specific targets for potential therapeutic modulation to improve short- and long-term respiratory health of preterm infants with BPD.


Asunto(s)
Displasia Broncopulmonar/genética , Corioamnionitis/patología , Hiperoxia/complicaciones , Pulmón/inmunología , Transcriptoma , Animales , Displasia Broncopulmonar/etiología , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro , Embarazo , Ratas , Ratas Sprague-Dawley
4.
Am J Med Genet A ; 182(1): 130-136, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31680412

RESUMEN

Costello syndrome (CS) is an autosomal-dominant condition caused by activating missense mutations in HRAS. There is little literature describing health concerns specific to adults with CS. Parents of individuals with CS need to know what to anticipate as their children age. We surveyed a group of 20 adults and older adolescents with CS regarding their medical concerns and lifestyle characteristics. We identified several previously undescribed actionable medical concerns in adults with CS. First, the high prevalence of anxiety in this cohort indicates that screening for anxiety is warranted since this is a treatable condition that can have a significant impact on quality of life. Second, adults with CS should be monitored for progressive contractures or other problems that could decrease mobility. This is especially important in a population that seems to have increased risk for osteopenia. Finally, the lack of cancer diagnoses in adulthood is of interest, although the cohort is too small to draw definitive conclusions about cancer risk in adults with CS. Ongoing follow-up of the current cohort of adults with CS is necessary to delineate progressive medical and physical problems, which is essential for providing targeted management recommendations and anticipatory guidance to families.


Asunto(s)
Ansiedad/epidemiología , Síndrome de Costello/epidemiología , Neoplasias/epidemiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Adolescente , Adulto , Ansiedad/complicaciones , Ansiedad/genética , Ansiedad/patología , Enfermedades Óseas Metabólicas/epidemiología , Enfermedades Óseas Metabólicas/genética , Enfermedades Óseas Metabólicas/patología , Niño , Síndrome de Costello/complicaciones , Síndrome de Costello/genética , Síndrome de Costello/patología , Femenino , Humanos , Masculino , Neoplasias/complicaciones , Neoplasias/genética , Neoplasias/patología , Fenotipo , Calidad de Vida , Adulto Joven
5.
Am J Hum Genet ; 96(5): 816-25, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25865493

RESUMEN

Transcription factors operate in developmental processes to mediate inductive events and cell competence, and perturbation of their function or regulation can dramatically affect morphogenesis, organogenesis, and growth. We report that a narrow spectrum of amino-acid substitutions within the transactivation domain of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a leucine zipper-containing transcription factor of the AP1 superfamily, profoundly affect development. Seven different de novo missense mutations involving conserved residues of the four GSK3 phosphorylation motifs were identified in eight unrelated individuals. The distinctive clinical phenotype, for which we propose the eponym Aymé-Gripp syndrome, is not limited to lens and eye defects as previously reported for MAF/Maf loss of function but includes sensorineural deafness, intellectual disability, seizures, brachycephaly, distinctive flat facial appearance, skeletal anomalies, mammary gland hypoplasia, and reduced growth. Disease-causing mutations were demonstrated to impair proper MAF phosphorylation, ubiquitination and proteasomal degradation, perturbed gene expression in primary skin fibroblasts, and induced neurodevelopmental defects in an in vivo model. Our findings nosologically and clinically delineate a previously poorly understood recognizable multisystem disorder, provide evidence for MAF governing a wider range of developmental programs than previously appreciated, and describe a novel instance of protein dosage effect severely perturbing development.


Asunto(s)
Catarata/genética , Sordera/genética , Glucógeno Sintasa Quinasa 3/genética , Discapacidad Intelectual/genética , Proteínas Proto-Oncogénicas c-maf/genética , Catarata/patología , Síndrome de Down/genética , Síndrome de Down/patología , Humanos , Discapacidad Intelectual/patología , Mutación , Fenotipo , Fosforilación , Convulsiones/genética , Convulsiones/patología
6.
Am J Med Genet A ; 173(5): 1309-1318, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28371260

RESUMEN

Costello syndrome is part of the RASopathies, a group of neurocardiofaciocutaneous syndromes caused by deregulation of the RAS mitogen-activated protein kinase pathway. Heterozygous mutations in HRAS are responsible for Costello syndrome, with more than 80% of the patients harboring the specific p.Gly12Ser variant. These individuals show a homogeneous phenotype. The clinical characteristics of the Costello syndrome individuals harboring rarer HRAS mutations are less understood, due to the small number of reported cases. Here, we describe the phenotypic spectrum of five additional individuals with HRAS c.38G>A; p.Gly13Asp, including one with somatic mosaicism, and review five previously described cases. The facial and hair abnormalities of the HRAS p.Gly13Asp individuals differ from the typical pattern observed in those showing the common HRAS (p.Gly12Ser) mutation, with less coarse facial features and slow growing, sparse hair with abnormal texture, the latter resembling the pattern observed in Noonan syndrome-like disorder with loose anagen hair and individuals harboring another amino acid substitution in HRAS (p.Gly13Cys). Although some individuals with HRAS p.Gly13Asp developed papillomata and vascular proliferation lesions, no malignant tumors occurred, similar to what was reported for individuals harboring the HRAS p.Gly13Cys. The fact that no malignant tumors were described in these individuals does not allow definitive conclusions about the risk for cancer development. It remains to be determined if substitutions of amino acid 13 in HRAS (p.Gly13Asp and p.Gly13Cys) increase the risk of tumor development.


Asunto(s)
Anomalías Múltiples/genética , Síndrome de Costello/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Anomalías Múltiples/fisiopatología , Adolescente , Adulto , Sustitución de Aminoácidos/genética , Aminoácidos/genética , Niño , Preescolar , Síndrome de Costello/complicaciones , Síndrome de Costello/fisiopatología , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Mosaicismo , Neoplasias/complicaciones , Neoplasias/fisiopatología , Fenotipo
7.
Am J Med Genet A ; 173(5): 1294-1300, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28374929

RESUMEN

Dysregulation of the mitogen activated protein kinase (MAPK) pathway in Costello syndrome (CS) may contribute to increased risk for autism-spectrum disorder (ASD). We examined prevalence of ASD symptoms in 14 individuals (six females) age 1-18 years with molecularly confirmed CS. Caregivers completed the Modified Checklist for Autism in Toddlers (M-CHAT) for ages 0-4 years (n = 7), and the Social Communication Questionnaire (SCQ) for ages 4 and older (n = 7). Age was associated with meeting ASD criteria: 5/7 (71.4%) younger children met the ASD cut-off on the MCHAT, compared to 0/7 older children on the SCQ. The following medical and developmental factors were strongly associated with ASD criteria on the M-CHAT: having a gastrostomy tube at time of assessment, not eating solid food, not walking, and not being toilet trained. Two children who met stricter ASD criteria had significantly lower adaptive functioning and were physically much more impaired. Among older participants, SCQ subscale scores in communication, socialization, and repetitive behavior domains were comparable to the typically-developing normative sample. ASD symptoms were highly elevated in younger CS individuals. Older children did not differ from typically developing samples in prevalence of ASD symptoms. CS individuals may appear to fall on the autism spectrum in early childhood due to severe feeding and orthopedic problems that improve by age four, suggesting many of these children may eventually emerge out of an ASD presentation.


Asunto(s)
Trastorno del Espectro Autista/epidemiología , Trastorno del Espectro Autista/fisiopatología , Síndrome de Costello/epidemiología , Síndrome de Costello/fisiopatología , Adolescente , Factores de Edad , Trastorno del Espectro Autista/genética , Niño , Preescolar , Síndrome de Costello/genética , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Conducta Social , Encuestas y Cuestionarios
8.
Am J Med Genet A ; 170(3): 559-64, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26572961

RESUMEN

Costello syndrome (CS) entails a cancer predisposition and is caused by activating HRAS mutations, typically arising de novo in the paternal germline. Hypoglycemia is common in CS neonates. A previously reported individual with the rare HRAS p.Gln22Lys had hyperinsulinemic hypoglycemia. Autopsy showed a discrete pancreatic nodule. The morphologic and immunohistochemistry findings, including loss of p57(Kip2) protein, were identical to a focal lesion of congenital hyperinsulinism, however, no KCNJ11 or ABCC8 mutation was identified and germline derived DNA showed no alternation of the maternal or paternal 11p15 alleles. Here we report paternal uniparental disomy (pUPD) within the lesion, similar to the pUPD11p15.5 in Beckwith-Wiedemann syndrome (BWS). The similar extent of the pUPD suggests a similar mechanism driving hyperinsulinemia in both conditions. After coincidental somatic LOH and pUPD, the growth promoting effects of the paternally derived HRAS mutation, in combination with the increased function of the adjacent paternally expressed IGF2, may together result in clonal expansion. Although this somatic LOH within pancreatic tissue resulted in hyperinsulinism, similar LOH in mesenchymal cells may drive embryonal rhabdomyosarcoma (ERMS). Interestingly, biallelic IGF2 expression has been linked to rhabdomyosarcoma tumorigenesis and pUPD11 occurred in all 8 ERMS samples from CS individuals. Somatic KRAS and HRAS mutations occur with comparable frequency in isolated malignancies. Yet, the malignancy risk in CS is notably higher than in Noonan syndrome with a KRAS mutation. It is conceivable that HRAS co-localization with IGF2 and the combined effect of pUPD 11p15.5 on both genes contributes to the oncogenic potential.


Asunto(s)
Síndrome de Beckwith-Wiedemann/genética , Hiperinsulinismo Congénito/genética , Síndrome de Costello/genética , Impresión Genómica , Hipoglucemia/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Disomía Uniparental/genética , Sustitución de Aminoácidos , Secuencia de Bases , Síndrome de Beckwith-Wiedemann/diagnóstico , Síndrome de Beckwith-Wiedemann/patología , Cromosomas Humanos Par 11/química , Células Clonales , Hiperinsulinismo Congénito/diagnóstico , Hiperinsulinismo Congénito/patología , Síndrome de Costello/diagnóstico , Síndrome de Costello/patología , Resultado Fatal , Humanos , Hipoglucemia/diagnóstico , Hipoglucemia/patología , Lactante , Patrón de Herencia , Factor II del Crecimiento Similar a la Insulina/genética , Pérdida de Heterocigocidad , Masculino , Datos de Secuencia Molecular , Páncreas/metabolismo , Páncreas/patología , Disomía Uniparental/diagnóstico , Disomía Uniparental/patología
9.
Am J Med Genet A ; 170(9): 2237-47, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27264673

RESUMEN

Noonan syndrome is a rasopathy caused by mutations in multiple genes encoding components of the RAS/MAPK pathway. Despite its variable phenotype, limited genotype-phenotype correlations exist. Noonan syndrome with loose anagen hair (NS-LAH) is characterized by its distinctive hair anomalies, developmental differences, and structural brain abnormalities and is caused by a single recurrent missense SHOC2 mutation. SHOC2 forms a complex with protein phosphatase 1 (PP1C). Protein phosphatases counterbalance kinases and control activation of signaling proteins, such as the mitogen-activated protein kinases of the RAS/MAPK pathway. Here we report four patients with de novo missense mutations in protein phosphatase one catalytic subunit beta (PPP1CB), sharing a recognizable phenotype. Three individuals had the recurrent PPP1CB c.146G>C, p.Pro49Arg mutation, the fourth had a c.166G>C, p.Ala56Pro change. All had relative or absolute macrocephaly, low-set and posteriorly angulated ears, and developmental delay. Slow growing and/or sparse hair and/or an unruly hair texture was present in all. Three individuals had feeding difficulties requiring feeding tubes. One of two males had cryptorchidism, another had pectus excavatum. Short stature was present in three. A female with the recurrent mutation had a Dandy-Walker malformation and optic nerve hypoplasia. Mild ventriculomegaly occurred in all, cerebellar tonsillar ectopia was seen in two and progressed to Chiari 1 malformation in one individual. Based on the combination of phenotypic findings and PPP1CB's effect on RAF dephosphorylation within the RAS/MAPK pathway, this novel condition can be considered a rasopathy, most similar to NS-LAH. Collectively, these mutations meet the standardized criteria for pathogenicity. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Síndrome del Cabello Anágeno Suelto/diagnóstico , Síndrome del Cabello Anágeno Suelto/genética , Mutación Missense , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Proteína Fosfatasa 1/genética , Encéfalo/patología , Niño , Preescolar , Síndrome de Dandy-Walker/diagnóstico , Síndrome de Dandy-Walker/genética , Diagnóstico por Imagen , Exoma , Facies , Femenino , Estudios de Asociación Genética , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndrome del Cabello Anágeno Suelto/metabolismo , Masculino , Síndrome de Noonan/metabolismo , Fenotipo , Adulto Joven , Proteínas ras/metabolismo
10.
Am J Med Genet A ; 170(12): 3197-3206, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27589201

RESUMEN

Costello syndrome (CS) arises from a typically paternally derived germline mutation in the proto-oncogene HRAS, and is considered a rasopathy. CS results in failure-to-thrive, intellectual disabilities, short stature, coarse facial features, skeletal abnormalities, congenital heart disease, and a predisposition for cancer, most commonly embryonal rhabdomyosarcoma (ERMS). The goal of this study was to characterize CS ERMS at the molecular level and to determine how divergent it is from sporadic ERMS. We characterized eleven ERMS tumors from eight unrelated CS patients, carrying paternally derived HRAS c.34G>A (p.Gly12Ser; 6) or c.35G>C (p.Gly12Ala; 2) mutations. Loss of heterozygosity (LOH) was evaluated in all CS ERMS by microarray and/or short tandem repeat (STR) markers spanning the entire chromosome 11. Eight CS ERMS tumors displayed complete paternal uniparental disomy of chromosome 11 (pUPD11), whereas two displayed UPD only at 11p and a second primary ERMS tumor showed UPD limited to 11p15.5, the classical hallmark for ERMS. Three sporadic ERMS cell lines (RD, Rh36, Rh18) and eight formalin fixed paraffin embedded (FFPE) ERMS tumors were also analyzed for RAS mutations and LOH status. We found a higher than anticipated frequency of RAS mutations (HRAS or NRAS; 50%) in sporadic ERMS cell lines/tumors. Unexpectedly, complete uniparental disomy (UPD11) was observed in five specimens, while the other six showed LOH extending across the p and q arms of chromosome 11. In this study, we are able to clearly demonstrate complete UPD11 in both syndromic and sporadic ERMS. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Síndrome de Costello/genética , Pérdida de Heterocigocidad/genética , Rabdomiosarcoma Embrionario/genética , Disomía Uniparental/genética , Adolescente , Niño , Preescolar , Cromosomas Humanos Par 11/genética , Síndrome de Costello/complicaciones , Síndrome de Costello/patología , Femenino , Genotipo , Mutación de Línea Germinal/genética , Humanos , Lactante , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas p21(ras)/genética , Rabdomiosarcoma Embrionario/etiología , Rabdomiosarcoma Embrionario/patología , Disomía Uniparental/patología
11.
BMC Urol ; 16(1): 62, 2016 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-27769252

RESUMEN

BACKGROUND: Copy number variation (CNV) is a potential contributing factor to many genetic diseases. Here we investigated the potential association of CNV with nonsyndromic cryptorchidism, the most common male congenital genitourinary defect, in a Caucasian population. METHODS: Genome wide genotyping were performed in 559 cases and 1772 controls (Group 1) using Illumina HumanHap550 v1, HumanHap550 v3 or Human610-Quad platforms and in 353 cases and 1149 controls (Group 2) using the Illumina Human OmniExpress 12v1 or Human OmniExpress 12v1-1. Signal intensity data including log R ratio (LRR) and B allele frequency (BAF) for each single nucleotide polymorphism (SNP) were used for CNV detection using PennCNV software. After sample quality control, gene- and CNV-based association tests were performed using cleaned data from Group 1 (493 cases and 1586 controls) and Group 2 (307 cases and 1102 controls) using ParseCNV software. Meta-analysis was performed using gene-based test results as input to identify significant genes, and CNVs in or around significant genes were identified in CNV-based association test results. Called CNVs passing quality control and signal intensity visualization examination were considered for validation using TaqMan CNV assays and QuantStudio® 3D Digital PCR System. RESULTS: The meta-analysis identified 373 genome wide significant (p < 5X10-4) genes/loci including 49 genes/loci with deletions and 324 with duplications. Among them, 17 genes with deletion and 1 gene with duplication were identified in CNV-based association results in both Group 1 and Group 2. Only 2 genes (NUCB2 and UPF2) containing deletions passed CNV quality control in both groups and signal intensity visualization examination, but laboratory validation failed to verify these deletions. CONCLUSIONS: Our data do not support that structural variation is a major cause of nonsyndromic cryptorchidism.


Asunto(s)
Criptorquidismo/genética , Variaciones en el Número de Copia de ADN , Población Blanca/genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Programas Informáticos
12.
Am J Med Genet A ; 167A(2): 271-81, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25394726

RESUMEN

Lateral meningocele syndrome (LMS, OMIM%130720), also known as Lehman syndrome, is a very rare skeletal disorder with facial anomalies, hypotonia and meningocele-related neurologic dysfunction. The characteristic lateral meningoceles represent the severe end of the dural ectasia spectrum and are typically most severe in the lower spine. Facial features of LMS include hypertelorism and telecanthus, high arched eyebrows, ptosis, midfacial hypoplasia, micrognathia, high and narrow palate, low-set ears and a hypotonic appearance. Hyperextensibility, hernias and scoliosis reflect a connective tissue abnormality, and aortic dilation, a high-pitched nasal voice, wormian bones and osteolysis may be present. Lateral meningocele syndrome has phenotypic overlap with Hajdu-Cheney syndrome. We performed exome resequencing in five unrelated individuals with LMS and identified heterozygous truncating NOTCH3 mutations. In an additional unrelated individual Sanger sequencing revealed a deleterious variant in the same exon 33. In total, five novel de novo NOTCH3 mutations were identified in six unrelated patients. One had a 26 bp deletion (c.6461_6486del, p.G2154fsTer78), two carried the same single base pair insertion (c.6692_93insC, p.P2231fsTer11), and three individuals had a nonsense point mutation at c.6247A > T (pK2083*), c.6663C > G (p.Y2221*) or c.6732C > A, (p.Y2244*). All mutations cluster into the last coding exon, resulting in premature termination of the protein and truncation of the negative regulatory proline-glutamate-serine-threonine rich PEST domain. Our results suggest that mutant mRNA products escape nonsense mediated decay. The truncated NOTCH3 may cause gain-of-function through decreased clearance of the active intracellular product, resembling NOTCH2 mutations in the clinically related Hajdu-Cheney syndrome and contrasting the NOTCH3 missense mutations causing CADASIL.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Exones , Meningocele/diagnóstico , Meningocele/genética , Mutación , Receptores Notch/genética , Niño , Preescolar , Análisis Mutacional de ADN , Exoma , Facies , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Imagen por Resonancia Magnética , Masculino , Fenotipo , Receptor Notch3 , Adulto Joven
13.
Am J Med Genet A ; 167A(9): 2085-97, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25914166

RESUMEN

Heterozygous germline mutations in the proto-oncogene HRAS cause Costello syndrome (CS), an intellectual disability condition with severe failure to thrive, cardiac abnormalities, predisposition to tumors, and neurologic abnormalities. More than 80% of patients share the HRAS mutation c.34G>A (p.Gly12Ser) associated with the typical, relatively homogeneous phenotype. Rarer mutations occurred in individuals with an attenuated phenotype and less characteristic facial features. Most pathogenic HRAS alterations affect hydrolytic HRAS activity resulting in constitutive activation. "Gain-of-function" and "hyperactivation" concerning downstream pathways are widely used to explain the molecular basis and dysregulation of the RAS-MAPK pathway is the biologic mechanism shared amongst rasopathies. Panel testing for rasopathies identified a novel HRAS mutation (c.179G>A; p.Gly60Asp) in three individuals with attenuated features of Costello syndrome. De novo paternal origin occurred in two, transmission from a heterozygous mother in the third. Individuals showed subtle facial features; curly hair and relative macrocephaly were seen in three; atrial tachycardia and learning difficulties in two, and pulmonic valve dysplasia and mildly thickened left ventricle in one. None had severe failure to thrive, intellectual disability or cancer, underscoring the need to consider HRAS mutations in individuals with an unspecific rasopathy phenotype. Functional studies revealed strongly increased HRAS(Gly60Asp) binding to RAF1, but not to other signaling effectors. Hyperactivation of the MAPK downstream signaling pathways was absent. Our results indicate that an increase in the proportion of activated RAS downstream signaling components does not entirely explain the molecular basis of CS. We conclude that the phenotypic variability in CS recapitulates variable qualities of molecular dysfunction.


Asunto(s)
Anomalías Múltiples/genética , Síndrome de Costello/genética , Predisposición Genética a la Enfermedad/genética , Mutación de Línea Germinal/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adolescente , Adulto , Niño , Femenino , Genes ras/genética , Humanos , Masculino , Proteínas Quinasas Activadas por Mitógenos/genética , Fenotipo , Proto-Oncogenes Mas , Transducción de Señal/genética
14.
Pediatr Blood Cancer ; 62(5): 751-8, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25728527

RESUMEN

BACKGROUND: Reovirus is a naturally occurring human virus that is cytopathic to malignant cells possessing an activated Ras signaling pathway. We conducted a phase I trial of Reolysin, a manufactured, proprietary isolate of purified reovirus, in children with relapsed/refractory extracranial solid tumors to define the recommended phase 2 dose (RP2D), toxicities, and pharmacokinetic properties when administered as a single agent or in combination with cyclophosphamide. PROCEDURES: Reolysin was administered intravenously for 5 consecutive days, every 28 days. Using a 3 + 3 design, the following dose levels were evaluated: 3 × 10(8) Tissue Culture Inhibitory Dose 50% (TCID50 )/kg; 5 × 10(8) TCID50 /kg (maximum dose was 3 × 10(10) TCID50 ); and 5 × 10(8) TCID50 /kg plus oral cyclophosphamide (50 mg/m(2) /day × 21 days). RESULTS: Twenty-nine patients were enrolled; 28 were eligible and 24 were evaluable for toxicity and response. There were no hematologic dose-limiting toxicities. Grade 5 respiratory failure and a Grade 5 thromboembolic event were reported, both in the setting of progressive disease. The median time to clear the reovirus viremia was 6.5 days. Eight of 24 patients were viremic beyond the 5 days of therapy, all were negative by day 17. No patient had detectable viral RNA in saliva or stool. There were no objective responses. CONCLUSIONS: Reolysin at a dose of 5 × 10(8) TCID50 /kg daily for 5 days was well tolerated in children alone and in combination with oral cyclophosphamide. Virus was cleared rapidly from the serum and shedding in stool and saliva was not detectable.


Asunto(s)
Ciclofosfamida/uso terapéutico , Resistencia a Antineoplásicos , Recurrencia Local de Neoplasia/terapia , Neoplasias/terapia , Viroterapia Oncolítica , Reoviridae , Terapia Recuperativa , Adolescente , Adulto , Antineoplásicos Alquilantes/uso terapéutico , Niño , Preescolar , Terapia Combinada , Femenino , Estudios de Seguimiento , Humanos , Masculino , Dosis Máxima Tolerada , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/virología , Estadificación de Neoplasias , Neoplasias/patología , Neoplasias/virología , Pronóstico , Adulto Joven
15.
Am J Med Genet A ; 164A(9): 2240-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24942156

RESUMEN

Patients with physical findings suggestive of Treacher Collins syndrome (TCS) or mandibulofacial dysostosis (MFD) and macrocytic anemia diagnostic of Diamond-Blackfan anemia (DBA) have been reported. Disease-causing genes have been identified for TCS and other MFDs. Mutations in several ribosomal protein genes and the transcription factor GATA1 result in DBA. However, no disease-causing mutation had been identified in the reported patients with the combination of TCS/MFD and DBA phenotype, and we hypothesized that pathogenic mutations in a single gene could be identified using whole exome analysis. We studied probands from six unrelated families. Combining exome analysis and Sanger sequencing, we identified likely pathogenic mutations in 5/6 families. Two mutations in unrelated families were seen in RPS26, the known DBA10 gene. One variant was predicted to affect mRNA splicing, and the other to lead to protein truncation. In another family a likely pathogenic X-linked mutation affecting a highly conserved residue was found in TSR2, which encodes a direct binding partner of RPS26. De novo mutations affecting the RPS28 start codon were found in two unrelated probands, identifying RPS28 as a novel disease gene. We conclude that the phenotype combining features of TCS with DBA is genetically heterogeneous. Each of the pathogenic variants identified is predicted to impede ribosome biogenesis, which in turn could result in altered cell growth and proliferation, causing abnormal embryologic development, defective erythropoiesis and reduced growth. The phenotype combining TCS/MFD and DBA is highly variable, overlaps with DBA and lies within the phenotypic spectrum of ribosomopathies. © 2014 Wiley Periodicals, Inc.


Asunto(s)
Anemia de Diamond-Blackfan/complicaciones , Anemia de Diamond-Blackfan/genética , Proteínas Reguladoras de la Apoptosis/genética , Heterogeneidad Genética , Disostosis Mandibulofacial/complicaciones , Disostosis Mandibulofacial/genética , Proteínas Ribosómicas/genética , Adulto , Preescolar , Exoma/genética , Familia , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Fenotipo , Embarazo , Adulto Joven
16.
Biol Reprod ; 89(1): 6, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23699388

RESUMEN

Plasma membrane Ca(2+)-ATPase isoform 4 (PMCA4) is the primary Ca(2+) efflux pump in murine sperm, where it regulates motility. In Pmca4 null sperm, motility loss results in infertility. We have shown that murine sperm PMCA4b interacts with Ca(2+)/CaM-dependent serine kinase (CASK) in regulating Ca(2+) homeostasis and motility. However, recent work indicated that the bovine PMCA4a splice variant (missing in testis) is epididymally expressed, along with 4b, and may be transferred to sperm. Here we show, via conventional and in situ RT-PCR, that both the splice variants of Pmca4 mRNA are expressed in murine testis and throughout the epididymis. Immunofluorescence localized PMCA4a to the apical membrane of the epididymal epithelium, and Western analysis not only confirmed its presence but showed for the first time that PMCA4a and PMCA4b are secreted in the epididymal luminal fluid (ELF), from which epididymosomes containing PMCA4a were isolated. Flow cytometry indicated the presence of PMCA4a on mature caudal sperm where it was increased ~5-fold compared to caput sperm (detected by Western blotting) and ~2-fold after incubation in ELF, revealing in vitro uptake and implicating PMCA4a in epididymal sperm maturation. Coimmunoprecipitation using pan-PMCA4 antibodies, revealed that both variants associate with CASK, suggesting their presence in a complex. Because they have different kinetic properties for Ca(2+) transport and different abilities to bind to CASK, our study suggests a mechanism for combining the functional attributes of both PMCA4 variants, leading to heightened efficiency of the pump in the maintenance of Ca(2+) homeostasis, which is crucial for normal motility and male fertility.


Asunto(s)
Epidídimo/enzimología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Maduración del Esperma , Espermatozoides/enzimología , Animales , Técnica del Anticuerpo Fluorescente , Guanilato-Quinasas , Inmunoprecipitación , Isoenzimas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Testículo/enzimología
17.
Genet Med ; 15(7): 554-7, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23429430

RESUMEN

PURPOSE: Costello syndrome, a rare genetic disorder with multisystemic involvement, is caused by germline HRAS mutations. Because several different missense mutations have been reported, a severity scoring system was developed to assess a possible genotype-phenotype correlation. METHODS: Records of 78 individuals with Costello syndrome were scored in early childhood, childhood, and young adulthood by a reviewer blinded to the individuals' specific mutations. These scores were based on certain medically relevant feeding, neurologic, orthopedic, endocrine, cardiac, malignancy, and mortality manifestations. Individuals' severity scores were then grouped by the particular HRAS mutation. The mixed-model approach for repeated-measures analysis of variance with unstructured within-subject correlation, pairwise comparisons, and contrast were used to determine whether the severity scores differed by mutation. RESULTS: Although the sample size was small, individuals with the p.G12A or p.G12C HRAS change were more severely affected than those with other HRAS mutations. Regardless of the mutation, severity did not increase significantly over time. CONCLUSION: Despite its limitations, including the small number of individuals with rare mutations and possibly incomplete medical records, this work providing the first quantitative assessment of phenotypic severity in a Costello syndrome cohort supports a medically relevant genotype-phenotype correlation.


Asunto(s)
Síndrome de Costello/etiología , Proteínas Proto-Oncogénicas p21(ras)/genética , Adolescente , Adulto , Factores de Edad , Niño , Preescolar , Síndrome de Costello/genética , Síndrome de Costello/mortalidad , Estudios de Asociación Genética , Humanos , Lactante , Mutación , Índice de Severidad de la Enfermedad , Adulto Joven
18.
Am J Med Genet A ; 161A(9): 2258-65, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23918324

RESUMEN

Costello syndrome (CS) is a rare genetic disorder caused by germline mutations in the HRAS proto-oncogene which belongs to the family of syndromes called rasopathies. HRAS plays a key role in synaptic long-term potentiation (LTP) and memory formation. Prior research has found impaired recall memory in CS despite enhancement in LTP that would predict memory preservation. Based on findings in other rasopathies, we hypothesized that the memory deficit in CS would be specific to recall, and that recognition memory would show relative preservation. Memory was tested using word-list learning and story memory tasks with both recall and recognition trials, a design that allowed us to examine these processes separately. Participants were 11 adolescents and young adults with molecularly confirmed CS, all of whom fell in the mild to moderate range of intellectual disability. Results indicated a clear dissociation between verbal recall, which was impaired (M = 69 ± 14), and recognition memory, which was relatively intact (M = 86 ± 14). Story recognition was highly correlated with listening comprehension (r = 0.986), which also fell in the low-average range (M = 80 ± 12.9). Performance on other measures of linguistic ability and academic skills was impaired. The findings suggest relatively preserved recognition memory that also provides some support for verbal comprehension. This is the first report of relatively normal performance in a cognitive domain in CS. Further research is needed to better understand the mechanisms by which altered RAS-MAPK signaling affects neuronal plasticity and memory processes in the brain.


Asunto(s)
Síndrome de Costello/psicología , Memoria , Aprendizaje Verbal , Adaptación Psicológica , Adolescente , Adulto , Femenino , Humanos , Pruebas del Lenguaje , Masculino , Proto-Oncogenes Mas , Reconocimiento en Psicología , Análisis y Desempeño de Tareas , Adulto Joven
19.
Am J Med Genet A ; 161A(10): 2420-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23918763

RESUMEN

Noonan syndrome is a heterogenous rasopathy typically presenting with short stature, characteristic facial features, cardiac abnormalities including pulmonic valve stenosis, ASD and hypertrophic cardiomyopathy (HCM), cryptorchidism, ectodermal abnormalities, and learning differences. The phenotype is variable, and limited genotype phenotype correlation exists with SOS1 mutations often associated with normal cognition and stature, RAF1 mutations entailing a high HCM risk, and certain PTPN11 mutations predisposing to juvenile myelomonocytic leukemia. The recently identified SHOC2 mutation (p.Ser2Gly) causes Noonan syndrome with loose anagen hair. We report five patients with this mutation. All had skin hyperpigmentation, sparse light colored hair, increased fine wrinkles, ligamentous laxity, developmental delay, and 4/4 had a structural cardiac anomaly. Hypotonia and macrocephaly occurred in 4/5 (80%); 3/5 (60%) had polyhydramnios, increased birth weight or required use of a feeding tube. Distinctive brain abnormalities included relative megalencephaly and enlarged subarachnoid spaces suggestive of benign external hydrocephalus, and a relatively small posterior fossa as indicated by a vertical tentorium. The combination of a large brain with a small posterior fossa likely resulted in the high rate of cerebellar tonsillar ectopia (3/4; 75%). Periventricular nodular heterotopia was seen in one patient with a thick and dysplastic corpus callosum. We report on the first hematologic neoplasm, myelofibrosis, in a 2-year-old patient with SHOC2 mutation. Myelofibrosis is exceedingly rare in children and young adults. The absence of a somatic JAK2 mutation, seen in the majority of patients with myelofibrosis, is noteworthy as it suggests that germline or somatic SHOC2 mutations are causally involved in myelofibrosis.


Asunto(s)
Cabello/patología , Péptidos y Proteínas de Señalización Intracelular/genética , Mutación , Síndrome de Noonan/diagnóstico , Síndrome de Noonan/genética , Fenotipo , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Adulto , Encéfalo/anomalías , Encéfalo/patología , Preescolar , Resultado Fatal , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Síndrome de Noonan/complicaciones , Mielofibrosis Primaria/complicaciones , Mielofibrosis Primaria/patología , Adulto Joven
20.
Appl Immunohistochem Mol Morphol ; 31(3): 154-162, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36735491

RESUMEN

The present study examined staining of guanylate cyclase C (GCC/GUCY2C) in the small and large intestines of children younger than age 7 years. Normal intestinal tissue from children aged 0 to 7 years was stained using GCC, uroguanylin, and villin antibodies and scored for staining intensity. A subset underwent quantitative real-time polymerase chain reaction. Data were analyzed using t test of independent means, descriptive statistics, and logistic regression. Four hundred sixty-four specimens underwent immunohistochemistry; 291 specimens underwent real-time polymerase chain reaction. GCC, villin, and uroguanylin were detected across age groups and anatomic sites. No significant differences were identifiable across age groups. GUCY2C and uroguanylin mRNA was detected in all samples, with no variability of statistical significance of either target-to-villin normalization between any age cohorts. A gradient of expression of GCC across age groups does not seem to exist.


Asunto(s)
Intestinos , Receptores Acoplados a la Guanilato-Ciclasa , Receptores de Péptidos , Niño , Preescolar , Humanos , Inmunohistoquímica , Microvellosidades/química , Microvellosidades/metabolismo , Receptores de Enterotoxina , Receptores Acoplados a la Guanilato-Ciclasa/genética , Receptores Acoplados a la Guanilato-Ciclasa/metabolismo , Receptores de Péptidos/genética , Receptores de Péptidos/metabolismo , Recién Nacido , Lactante
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA