Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Genet Metab ; 140(3): 107696, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37690181

RESUMEN

PURPOSE: Individuals with urea cycle disorders (UCDs) may develop recurrent hyperammonemia, episodic encephalopathy, and neurological sequelae which can impact Health-related Quality of Life (HRQoL). To date, there have been no systematic studies of HRQoL in people with UCDs. METHODS: We reviewed HRQoL and clinical data for 190 children and 203 adults enrolled in a multicenter UCD natural history study. Physical and psychosocial HRQoL in people with UCDs were compared to HRQoL in healthy people and people with phenylketonuria (PKU) and diabetes mellitus. We assessed relationships between HRQoL, UCD diagnosis, and disease severity. Finally, we calculated sample sizes required to detect changes in these HRQoL measures. RESULTS: Individuals with UCDs demonstrated worse physical and psychosocial HRQoL than their healthy peers and peers with PKU and diabetes. In children, HRQoL scores did not differ by diagnosis or severity. In adults, individuals with decreased severity had worse psychosocial HRQoL. Finally, we show that a large number of individuals would be required in clinical trials to detect differences in HRQoL in UCDs. CONCLUSION: Individuals with UCDs have worse HRQoL compared to healthy individuals and those with PKU and diabetes. Future work should focus on the impact of liver transplantation and other clinical variables on HRQoL in UCDs.


Asunto(s)
Diabetes Mellitus , Hiperamonemia , Trasplante de Hígado , Fenilcetonurias , Trastornos Innatos del Ciclo de la Urea , Niño , Humanos , Adulto , Calidad de Vida , Trastornos Innatos del Ciclo de la Urea/diagnóstico , Hiperamonemia/diagnóstico , Fenilcetonurias/complicaciones , Estudios Multicéntricos como Asunto
2.
ERJ Open Res ; 7(2)2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34104643

RESUMEN

BACKGROUND: Healthcare workers (HCWs) are believed to be at increased risk of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. It is not known to what extent the natural production of antibodies to SARS-CoV-2 is protective against re-infection. METHODS: A prospective observational study of HCWs in Scotland (UK) from May to September 2020 was performed. The Siemens SARS-CoV-2 total antibody assay was used to establish seroprevalence in this cohort. Controls, matched for age and sex to the general local population, were studied for comparison. New infections (up to 2 December 2020) post antibody testing were recorded to determine whether the presence of SARS-CoV-2 antibodies protects against re-infection. RESULTS: A total of 2063 health and social care workers were recruited for this study. At enrolment, 300 HCWs had a positive antibody test (14.5%). 11 out of 231 control sera tested positive (4.8%). HCWs therefore had an increased likelihood of a positive test (OR 3.4, 95% CI 1.85-6.16; p<0.0001). Dentists were most likely to test positive. 97.3% of patients who had previously tested positive for SARS-CoV-2 by reverse transcriptase (RT)-PCR had positive antibodies. 18.7% had an asymptomatic infection. There were 38 new infections with SARS-CoV-2 in HCWs who were previously antibody negative, and one symptomatic RT-PCR-positive re-infection. The presence of antibodies was therefore associated with an 85% reduced risk of re-infection with SARS-CoV-2 (hazard ratio 0.15, 95% CI 0.06-0.35; p=0.026). CONCLUSION: HCWs were three times more likely to test positive for SARS-CoV-2 than the general population. Almost all infected individuals developed an antibody response, which was 85% effective in protecting against re-infection with SARS-CoV-2.

3.
J Pediatr Hematol Oncol ; 42(2): e114-e116, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-30789458

RESUMEN

Idiopathic hyperammonemia is a rare, poorly understood, and often lethal condition that has been described in immunocompromised patients. This report describes an immunocompromised patient with acute myelogenous leukemia who developed persistent hyperammonemia up to 705 µmol/L (normal, 0 to 47 µmol/L) refractory to multiple different therapies. However, after beginning azithromycin and then doxycycline therapy for Ureaplasma species infection, the patient showed immediate and sustained clinical improvement and resolution of ammonia levels. Recognizing disseminated Ureaplasma species infection as a potential cause of idiopathic hyperammonemia, an unexplained, often fatal condition in immunocompromised patients, and empirically treating for this infection could potentially be lifesaving.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Hiperamonemia/etiología , Huésped Inmunocomprometido/efectos de los fármacos , Quimioterapia de Inducción/efectos adversos , Leucemia Mieloide Aguda/tratamiento farmacológico , Infecciones por Ureaplasma/complicaciones , Ureaplasma/efectos de los fármacos , Adolescente , Antibacterianos/uso terapéutico , Doxiciclina/uso terapéutico , Femenino , Humanos , Hiperamonemia/tratamiento farmacológico , Hiperamonemia/patología , Leucemia Mieloide Aguda/microbiología , Leucemia Mieloide Aguda/patología , Pronóstico , Infecciones por Ureaplasma/inducido químicamente , Infecciones por Ureaplasma/microbiología
4.
Genet Med ; 22(3): 490-499, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31607746

RESUMEN

PURPOSE: We investigated the value of transcriptome sequencing (RNAseq) in ascertaining the consequence of DNA variants on RNA transcripts to improve the diagnostic rate from exome or genome sequencing for undiagnosed Mendelian diseases spanning a wide spectrum of clinical indications. METHODS: From 234 subjects referred to the Undiagnosed Diseases Network, University of California-Los Angeles clinical site between July 2014 and August 2018, 113 were enrolled for high likelihood of having rare undiagnosed, suspected genetic conditions despite thorough prior clinical evaluation. Exome or genome sequencing and RNAseq were performed, and RNAseq data was integrated with genome sequencing data for DNA variant interpretation genome-wide. RESULTS: The molecular diagnostic rate by exome or genome sequencing was 31%. Integration of RNAseq with genome sequencing resulted in an additional seven cases with clear diagnosis of a known genetic disease. Thus, the overall molecular diagnostic rate was 38%, and 18% of all genetic diagnoses returned required RNAseq to determine variant causality. CONCLUSION: In this rare disease cohort with a wide spectrum of undiagnosed, suspected genetic conditions, RNAseq analysis increased the molecular diagnostic rate above that possible with genome sequencing analysis alone even without availability of the most appropriate tissue type to assess.


Asunto(s)
Enfermedades Genéticas Congénitas/diagnóstico , Patología Molecular , Enfermedades Raras/diagnóstico , Transcriptoma/genética , Exoma/genética , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas/normas , Humanos , Mutación/genética , RNA-Seq/normas , Enfermedades Raras/genética , Análisis de Secuencia de ADN/normas , Secuenciación del Exoma/normas , Secuenciación Completa del Genoma/normas
5.
Mol Genet Genomic Med ; 8(2): e1027, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31872981

RESUMEN

BACKGROUND: When time is of the essence in critical care cases, a fast molecular diagnosis is often necessary to help health care providers quickly determine best next steps for treatments, prognosis, and counseling of their patients. In this paper, we present the diagnostic rates and improved quality of life for patients undergoing clinical rapid exome sequencing. METHODS: The clinical histories and results of 41 patients undergoing rapid exome sequencing were retrospectively reviewed. RESULTS: Clinical rapid exome sequencing identified a definitive diagnosis in 13/41 (31.7%) and other relevant findings in 17 of the patients (41.5%). The average time to verbal report was 7 days; to written report was 11 days. CONCLUSIONS: Our observations demonstrate the utility and effectiveness of rapid family-based diagnostic exome sequencing in improving patients care.


Asunto(s)
Secuenciación del Exoma/normas , Pruebas Genéticas/normas , Adolescente , Adulto , Niño , Preescolar , Diagnóstico Precoz , Femenino , Pruebas Genéticas/métodos , Humanos , Lactante , Masculino , Secuenciación del Exoma/métodos
6.
Am J Med Genet A ; 179(3): 498-502, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30676690

RESUMEN

ALG11-Congenital Disorder of Glycosylation (ALG11-CDG, also known as congenital disorder of glycosylation type Ip) is an inherited inborn error of metabolism due to abnormal protein and lipid glycosylation. We describe two unrelated patients with ALG11-CDG due to novel mutations, review the literature of previously described affected individuals, and further expand the clinical phenotype. Both affected individuals reported here had severe psychomotor disabilities and epilepsy. Their fibroblasts synthesized truncated precursor glycan structures, consistent with ALG11-CDG, while also showing hypoglycosylation of a novel biomarker, GP130. Surprisingly, one patient presented with normal transferrin glycosylation profile, a feature that has not been reported previously in patients with ALG11-CDG. Together, our data expand the clinical and mutational spectrum of ALG11-CDG.


Asunto(s)
Trastornos Congénitos de Glicosilación/diagnóstico , Trastornos Congénitos de Glicosilación/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Manosiltransferasas/genética , Mutación , Fenotipo , Adolescente , Alelos , Biomarcadores , Preescolar , Electroencefalografía , Femenino , Glicosilación , Humanos , Imagen por Resonancia Magnética , Masculino , Linaje , Tomografía Computarizada por Rayos X
7.
BMJ Case Rep ; 20182018 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-29386211

RESUMEN

Neurofibromatosis type 1 (NF1)-related lung disease is a rare but increasingly recognised, high morbidity associated feature of the condition. We present a 48-year-old male patient with NF1, who was initially admitted for a subarachnoid haemorrhage requiring aneurysmal coil embolisation. During his recovery, he developed a left-sided pneumothorax requiring chest tube placement followed by concerns for re-expansion pulmonary oedema requiring intubation. Subsequently, the patient also developed a right-sided pneumothorax requiring additional chest tube placement but did not develop right-sided pulmonary oedema. During his hospitalisation, the patient also exemplified other important NF1-related pathophysiology including pheochromocytoma, cerebrovascular abnormalities and cardiovascular manifestations. Due to his multiple comorbidities and poor prognosis, we held a goals of care discussion with the patient's mother, and with her agreement, the patient underwent compassionate withdrawal of artificial life support.


Asunto(s)
Cuidados para Prolongación de la Vida , Pulmón/diagnóstico por imagen , Neurofibromatosis 1/fisiopatología , Neumotórax/diagnóstico por imagen , Edema Pulmonar/diagnóstico por imagen , Accidente Cerebrovascular/fisiopatología , Hemorragia Subaracnoidea/fisiopatología , Privación de Tratamiento , Neoplasias de las Glándulas Suprarrenales , Tubos Torácicos , Comorbilidad , Toma de Decisiones , Progresión de la Enfermedad , Embolización Terapéutica , Humanos , Intubación , Masculino , Persona de Mediana Edad , Neurofibromatosis 1/complicaciones , Neurofibromatosis 1/terapia , Feocromocitoma , Neumotórax/fisiopatología , Neumotórax/terapia , Pronóstico , Edema Pulmonar/fisiopatología , Edema Pulmonar/terapia , Accidente Cerebrovascular/diagnóstico por imagen , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/terapia , Hemorragia Subaracnoidea/diagnóstico por imagen , Hemorragia Subaracnoidea/terapia , Cuidado Terminal
8.
Am J Med Genet A ; 173(5): 1319-1327, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28296084

RESUMEN

The cohesin complex is an evolutionarily conserved multi-subunit protein complex which regulates sister chromatid cohesion during mitosis and meiosis. Additionally, the cohesin complex regulates DNA replication, DNA repair, and transcription. The core of the complex consists of four subunits: SMC1A, SMC3, RAD21, and STAG1/2. Loss-of-function mutations in many of these proteins have been implicated in human developmental disorders collectively termed "cohesinopathies." Through clinical exome sequencing (CES) of an 8-year-old girl with a clinical history of global developmental delay, microcephaly, microtia with hearing loss, language delay, ADHD, and dysmorphic features, we describe a heterozygous de novo variant (c.205C>T; p.(Arg69*)) in the integral cohesin structural protein, STAG2. This variant is associated with decreased STAG2 protein expression. The analyses of metaphase spreads did not exhibit premature sister chromatid separation; however, delayed sister chromatid cohesion was observed. To further support the pathogenicity of STAG2 variants, we identified two additional female cases from the DECIPHER research database with mutations in STAG2 and phenotypes similar to our patient. Interestingly, the clinical features of these three cases are remarkably similar to those observed in other well-established cohesinopathies. Herein, we suggest that STAG2 is a dosage-sensitive gene and that heterozygous loss-of-function variants lead to a cohesinopathy.


Asunto(s)
Antígenos Nucleares/genética , Anomalías Congénitas/genética , Discapacidades del Desarrollo/genética , Microcefalia/genética , Antígenos Nucleares/biosíntesis , Proteínas de Ciclo Celular/genética , Niño , Proteínas Cromosómicas no Histona/genética , Anomalías Congénitas/fisiopatología , Discapacidades del Desarrollo/fisiopatología , Femenino , Regulación de la Expresión Génica , Heterocigoto , Humanos , Microcefalia/fisiopatología , Cohesinas
9.
Genet Med ; 18(2): 162-7, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25856670

RESUMEN

PURPOSE: We evaluated the clinical outcome in homocysteine remethylation disorders following newborn screening (NBS) and initiation of early specific treatment. METHODS: Five patients with remethylation disorders were included in this study. RESULTS: Two asymptomatic patients (one with cblG and one with cblE) were identified by NBS using an approach that combines a postanalytical interpretive tool (available on the Region 4 Stork (R4S) collaborative project website, http://www.clir-r4s.org) and a second-tier test for total homocysteine determination. Both the initial screening and the second-tier test are performed on the same blood spot, with no additional patient contact, resulting in no false-positive outcomes. Two additional patients with methylenetetrahydrofolate reductase deficiency were detected by NBS using low methionine as a marker. Although already symptomatic despite the early diagnosis, the latter two patients greatly improved with treatment and their outcomes are compared with that of another patient with methylenetetrahydrofolate reductase deficiency and significant morbidity who was diagnosed clinically at 3 months of age. CONCLUSION: Early detection by NBS and timely and specific treatment considerably improve at least short-term outcomes of homocysteine remethylation disorders. When a remethylation disorder is suspected, group-specific treatment could be started prior to the completion of in vitro confirmatory testing because all disorders from this group require similar intervention.


Asunto(s)
Errores Innatos del Metabolismo de los Aminoácidos/diagnóstico , Errores Innatos del Metabolismo de los Aminoácidos/terapia , Homocisteína/metabolismo , Tamizaje Neonatal , Femenino , Homocistinuria/diagnóstico , Humanos , Recién Nacido , Masculino , Metionina/metabolismo , Metilenotetrahidrofolato Reductasa (NADPH2)/deficiencia , Espasticidad Muscular/diagnóstico , Trastornos Psicóticos/diagnóstico , Resultado del Tratamiento
10.
Eur J Hum Genet ; 23(11): 1473-81, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25944381

RESUMEN

Dual-specificity tyrosine-(Y)-phosphorylation-regulated kinase 1 A (DYRK1A ) is a highly conserved gene located in the Down syndrome critical region. It has an important role in early development and regulation of neuronal proliferation. Microdeletions of chromosome 21q22.12q22.3 that include DYRK1A (21q22.13) are rare and only a few pathogenic single-nucleotide variants (SNVs) in the DYRK1A gene have been described, so as of yet, the landscape of DYRK1A disruptions and their associated phenotype has not been fully explored. We have identified 14 individuals with de novo heterozygous variants of DYRK1A; five with microdeletions, three with small insertions or deletions (INDELs) and six with deleterious SNVs. The analysis of our cohort and comparison with published cases reveals that phenotypes are consistent among individuals with the 21q22.12q22.3 microdeletion and those with translocation, SNVs, or INDELs within DYRK1A. All individuals shared congenital microcephaly at birth, intellectual disability, developmental delay, severe speech impairment, short stature, and distinct facial features. The severity of the microcephaly varied from -2 SD to -5 SD. Seizures, structural brain abnormalities, eye defects, ataxia/broad-based gait, intrauterine growth restriction, minor skeletal abnormalities, and feeding difficulties were present in two-thirds of all affected individuals. Our study demonstrates that haploinsufficiency of DYRK1A results in a new recognizable syndrome, which should be considered in individuals with Angelman syndrome-like features and distinct facial features. Our report represents the largest cohort of individuals with DYRK1A disruptions to date, and is the first attempt to define consistent genotype-phenotype correlations among subjects with 21q22.13 microdeletions and DYRK1A SNVs or small INDELs.


Asunto(s)
Síndrome de Down/genética , Discapacidad Intelectual/genética , Microcefalia/genética , Proteínas Serina-Treonina Quinasas/genética , Proteínas Tirosina Quinasas/genética , Anomalías Múltiples/genética , Anomalías Múltiples/fisiopatología , Deleción Cromosómica , Síndrome de Down/patología , Facies , Femenino , Haploinsuficiencia , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Microcefalia/fisiopatología , Fenotipo , Polimorfismo de Nucleótido Simple , Quinasas DyrK
11.
Gene ; 556(2): 249-53, 2015 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-25434494

RESUMEN

Ornithine transcarbamylase deficiency (OTCD, OMIM 311250), the most common urea cycle disorder, results in impaired synthesis of citrulline from carbamoyl phosphate and ornithine. Individuals have been identified with OTCD due to a contiguous gene deletion at Xp11.4-p21.1 and unique clinical features, described as the "extended OTCD phenotype". We present a male with neonatal-lethal OTCD due to a 1.87Mb microdeletion at Xp11.4-p21.1 (37126841-38998991 hg18). Autopsy revealed a novel histological finding of hepatocyte globular and granular inclusions. Such inclusions have not been described in OTCD or other metabolic disorders and are not an associated finding in neonatal liver failure due to other causes. The deleted region includes the gene SYTL5, potentially involved in RAB27A-dependent membrane trafficking in the liver and placenta. We propose that the contiguous gene deletion could contribute to the severity of the clinical presentation here and hypothesize that deletion of SYTL5 could contribute to the liver findings.


Asunto(s)
Proteínas Portadoras/genética , Deleción Cromosómica , Hígado/patología , Proteínas de la Membrana/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/patología , Cromosomas Humanos X , Humanos , Recién Nacido , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Polimorfismo de Nucleótido Simple
12.
Mol Genet Metab ; 110(4): 477-83, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24103308

RESUMEN

BACKGROUND AND METHODS: There are considerable uncertainty and debate regarding all aspects of newborn screen-positive cases of 3-methylcrotonyl-CoA carboxylase deficiency (3-MCCD), including diagnostic criteria, clinical spectrum, morbidity, prognosis, and appropriate management. To address some of these questions, we queried data from the California Newborn Screening Program's Screening Information System (SIS) and available scanned laboratory reports on cases of 3-MCCD reported by 15 state contracted metabolic specialty care centers born between July 2005 and December 2010. We evaluated the completeness and utility of the database as a tool for clinical disease characterization. RESULTS: During the study period, 2,959,108 infants were screened and 71 infants were diagnosed with 3-MCCD for an overall incidence of 1:41,676. The availability of diagnostic biochemical laboratory data varied significantly from subject to subject. Using a new case classification based on biochemical severity, we found that 8 of the cases met our criteria for biochemically severe (category 1), 19 cases met our criteria for biochemically mild (category 2) that we suspect to possibly be hypomorphic variants or heterozygote carriers, and 44 cases could not be classified (category 3) as mild or severe based on the data available in SIS. Documentation of the treatment regimens also varied significantly with 49% receiving dietary modification and 44% receiving carnitine. 15% of cases were documented to have experienced at least one of the following symptoms: lethargy, vomiting, irritability, ketosis, poor feeding, or poor tone. The majority of the subjects were completely developmentally age appropriate at their last assessment. CONCLUSIONS: The results suggest that a significant portion of the 3-MCCD "confirmed" cases have a mild biochemical phenotype. Moreover the majority of cases had insufficient data entered to allow for adequate clinical characterization of the cases. These findings raise the concern that a significant number of individuals receiving treatment for 3-MCCD may not have a clinically significant condition. Additionally, the utility of this data system could be improved if centers provided complete confirmatory test results and more specific documentation of clinical outcomes and health/developmental status. Further studies, including a clinical chart review, are necessary to validate the data and further characterize this cohort.


Asunto(s)
Ligasas de Carbono-Carbono/deficiencia , Tamizaje Neonatal , Trastornos Innatos del Ciclo de la Urea/genética , Acetonitrilos , California , Ligasas de Carbono-Carbono/genética , Carnitina , Humanos , Lactante , Recién Nacido , Trastornos Innatos del Ciclo de la Urea/epidemiología , Trastornos Innatos del Ciclo de la Urea/patología
13.
JIMD Rep ; 11: 99-106, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23606313

RESUMEN

OBJECTIVE: Barth syndrome is an X-linked recessive disorder characterized by dilated cardiomyopathy, neutropenia, 3-methylglutaconic aciduria, abnormal mitochondria, variably expressed skeletal myopathy, and growth delay. The disorder is caused by mutations in the tafazzin (TAZ/G4.5) gene located on Xq28. We report a novel exonic splicing mutation in the TAZ gene in a patient with atypical Barth syndrome. PATIENT & METHODS: The 4-month-old proband presented with respiratory distress, neutropenia, and dilated cardiomyopathy with reduced ejection fraction of 10%. No 3-methylglutaconic aciduria was detected on repeated urine organic acid analyses. Family history indicated that his maternal uncle died of endocardial fibroelastosis and dilated cardiomyopathy at 26 months. TAZ DNA sequencing, mRNA analysis, and cardiolipin analysis were performed. RESULTS: A novel nucleotide substitution c.553A>G in exon 7 of the TAZ gene was identified in the proband, predicting an amino acid substitution p.Met185Val. However, this mutation created a new splice donor signal within exon 7 causing mis-splicing of the message, producing two messages that only differ in the presence/absence of exon 5; these retain intron 6 and have only 11 bases of exon 7. Cardiolipin analysis confirmed the loss of tafazzin activity. The proband's mother, maternal aunt, and grandmother carry the same mutation. CONCLUSIONS: The identification of a TAZ gene mutation, mRNA analysis, and monolysocardiolipin/cardiolipin ratio determination were important for the diagnosis and genetic counseling in this family with atypical Barth syndrome that was not found to be associated with 3-methylglutaconic aciduria.

14.
Mol Genet Metab ; 106(1): 55-61, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22424739

RESUMEN

BACKGROUND: Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is an autosomal recessive inborn error of mitochondrial fatty acid oxidation with highly variable biochemical, genetic, and clinical characteristics. SCADD has been associated with accumulation of butyryl-CoA byproducts, including butyrylcarnitine (C4), butyrylglycine, ethylmalonic acid (EMA), and methylsuccinic acid (MS) in body fluid and tissues. Differences in genotype frequencies have been shown between patients diagnosed clinically versus those diagnosed by newborn screening. Moreover, while patients diagnosed clinically have a variable clinical presentation including developmental delay, ketotic hypoglycemia, epilepsy and behavioral disorders, studies suggest patients diagnosed by newborn screening are largely asymptomatic. Scant information is published about the biochemical, genetic and clinical outcome of SCADD patients diagnosed by newborn screening. METHODS: We collected California newborn screening, follow-up biochemical levels, and ACADS mutation data from September, 2005 through April, 2010. We retrospectively reviewed available data on SCADD cases diagnosed by newborn screening for clinical outcomes. RESULTS: During the study period, 2,632,058 newborns were screened and 76 confirmed SCADD cases were identified. No correlations between initial C4 value and follow-up biochemical markers (C4, EMA or MS levels) were found in the 76 cases studied. We found significant correlation between urine EMA versus MS, and correlation between follow-up C4 versus urine EMA. Of 22 cases where ACADS gene sequencing was performed: 7 had two or more deleterious mutations; 8 were compound heterozygotes for a deleterious mutation and common variant; 7 were homozygous for the common variant c.625G>A; and 1 was heterozygous for c.625G>A. Significant increases in mean urine EMA and MS levels were noted in patients with two or more deleterious mutations versus mutation heterozygotes or common polymorphism homozygotes. Clinical outcome data was available in 31 patients with follow-up extending from 0.5 to 60 months. None developed epilepsy or behavioral disorders, and three patients had isolated speech delay. Hypoglycemia occurred in two patients, both in the neonatal period. The first patient had concomitant meconium aspiration; the other presented with central apnea, poor feeding, and hypotonia. The latter, a c.625G>A homozygote, has had persistent elevations in both short- and medium-chain acylcarnitines; diagnostic workup in this case is extensive and ongoing. CONCLUSIONS: This study examines the largest series to date of SCADD patients identified by newborn screening. Our results suggest that confirmatory tests may be useful to differentiate patients with common variants from those with deleterious mutations. This study also provides evidence to suggest that, even when associated with deleterious mutations, SCADD diagnosed by newborn screening presents largely as a benign condition.


Asunto(s)
Acilcoenzima A , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/genética , Tamizaje Neonatal , Acilcoenzima A/sangre , Acilcoenzima A/genética , Acilcoenzima A/orina , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , California , Carnitina/análogos & derivados , Carnitina/sangre , Carnitina/orina , Femenino , Estudios de Seguimiento , Humanos , Recién Nacido , Masculino , Malonatos/sangre , Malonatos/orina , Eliminación de Secuencia , Succinatos/sangre , Succinatos/orina
15.
Am J Med Genet A ; 155A(11): 2871-8, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21994138

RESUMEN

The pontocerebellar hypoplasias (PCHs) are a group of clinically variable disorders characterized by abnormally small cerebellum and brainstem, generally inherited in an autosomal recessive pattern. While PCHs have been grouped into six subtypes, clinical diagnosis is equivocal until a genetic diagnosis is established. We report a patient with PCH, intrauterine growth restriction, ventricular septal defect, rib anomalies, midgut malrotation, and facial dysmorphic features. Using SNP analysis, we identified three de novo deletions of: 1.055 Mb at 6q24.3q25.1 (148174730-149229583); 169 kb at 16p13.2 (6565411-6733934); and 2.530 Mb at 19p13.11p13.12 (13857587-16387798), which were confirmed by FISH. 19p13 deletions are rare aberrations. Of patients previously described with overlapping 19p13.12 deletions and multiple anomalies, only one presented with PCH. Deleted in both that patient and the patient reported here, is DDX39, a DEAD-box RNA helicase. DDX39 is part of the homeostatic machinery that regulates the switch of cellular proliferation and differentiation. It is highly expressed in the developing central nervous system and optic cup of Xenopus laevis. The brain abnormalities in the patient reported here were more severe than the previously reported patient, which may be due to additional deletions or undetected point mutations in the nondeleted allele. Notably, the patient reported here also has a partial deletion of RBFOX1 (A2BP1), which lies within the autism susceptibility locus on 16p13.2. Our findings suggest chromosomal microarray analysis may be useful in determining etiology of syndromic PCH.


Asunto(s)
Anomalías Múltiples/genética , Deleción Cromosómica , Cromosomas Humanos Par 19/genética , Atrofias Olivopontocerebelosas/genética , Anomalías Múltiples/diagnóstico , Cromosomas Humanos Par 16/genética , Cromosomas Humanos Par 6/genética , ARN Helicasas DEAD-box/genética , Resultado Fatal , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Atrofias Olivopontocerebelosas/diagnóstico , Atrofias Olivopontocerebelosas/patología , Mutación Puntual , Polimorfismo de Nucleótido Simple , Embarazo , Complicaciones del Embarazo/patología , Factores de Empalme de ARN , Proteínas de Unión al ARN/genética
16.
Dev Cell ; 17(1): 49-61, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19619491

RESUMEN

Wnt/beta-catenin and NF-kappaB signaling mechanisms provide central controls in development and disease, but how these pathways intersect is unclear. Using hair follicle induction as a model system, we show that patterning of dermal Wnt/beta-catenin signaling requires epithelial beta-catenin activity. We find that Wnt/beta-catenin signaling is absolutely required for NF-kappaB activation, and that Edar is a direct Wnt target gene. Wnt/beta-catenin signaling is initially activated independently of EDA/EDAR/NF-kappaB activity in primary hair follicle primordia. However, Eda/Edar/NF-kappaB signaling is required to refine the pattern of Wnt/beta-catenin activity, and to maintain this activity at later stages of placode development. We show that maintenance of localized expression of Wnt10b and Wnt10a requires NF-kappaB signaling, providing a molecular explanation for the latter observation, and identify Wnt10b as a direct NF-kappaB target. These data reveal a complex interplay and interdependence of Wnt/beta-catenin and EDA/EDAR/NF-kappaB signaling pathways in initiation and maintenance of primary hair follicle placodes.


Asunto(s)
Ectodisplasinas/metabolismo , Folículo Piloso/embriología , FN-kappa B/metabolismo , Receptores de la Ectodisplasina/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Diferenciación Celular/fisiología , Ectodermo/citología , Ectodermo/metabolismo , Ectodisplasinas/genética , Embrión de Mamíferos/anatomía & histología , Embrión de Mamíferos/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Folículo Piloso/citología , Folículo Piloso/fisiología , Ratones , Ratones Transgénicos , FN-kappa B/genética , Embarazo , Receptores de la Ectodisplasina/genética , Piel/citología , Piel/embriología , Piel/metabolismo , Proteínas Wnt/genética , beta Catenina/genética
17.
Dev Biol ; 313(1): 210-24, 2008 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-18022614

RESUMEN

Wnt/beta-catenin signaling plays key roles in tooth development, but how this pathway intersects with the complex interplay of signaling factors regulating dental morphogenesis has been unclear. We demonstrate that Wnt/beta-catenin signaling is active at multiple stages of tooth development. Mutation of beta-catenin to a constitutively active form in oral epithelium causes formation of large, misshapen tooth buds and ectopic teeth, and expanded expression of signaling molecules important for tooth development. Conversely, expression of key morphogenetic regulators including Bmp4, Msx1, and Msx2 is downregulated in embryos expressing the secreted Wnt inhibitor Dkk1 which blocks signaling in epithelial and underlying mesenchymal cells. Similar phenotypes are observed in embryos lacking epithelial beta-catenin, demonstrating a requirement for Wnt signaling within the epithelium. Inducible Dkk1 expression after the bud stage causes formation of blunted molar cusps, downregulation of the enamel knot marker p21, and loss of restricted ectodin expression, revealing requirements for Wnt activity in maintaining secondary enamel knots. These data place Wnt/beta-catenin signaling upstream of key morphogenetic signaling pathways at multiple stages of tooth development and indicate that tight regulation of this pathway is essential both for patterning tooth development in the dental lamina, and for controlling the shape of individual teeth.


Asunto(s)
Morfogénesis , Diente/embriología , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Ratones , Odontogénesis , Transducción de Señal , Diente/metabolismo
18.
Nat Genet ; 39(1): 106-12, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17128274

RESUMEN

Fungiform taste papillae form a regular array on the dorsal tongue. Taste buds arise from papilla epithelium and, unusually for epithelial derivatives, synapse with neurons, release neurotransmitters and generate receptor and action potentials. Despite the importance of taste as one of our five senses, genetic analyses of taste papilla and bud development are lacking. We demonstrate that Wnt-beta-catenin signaling is activated in developing fungiform placodes and taste bud cells. A dominant stabilizing mutation of epithelial beta-catenin causes massive overproduction of enlarged fungiform papillae and taste buds. Likewise, genetic deletion of epithelial beta-catenin or inhibition of Wnt-beta-catenin signaling by ectopic dickkopf1 (Dkk1) blocks initiation of fungiform papilla morphogenesis. Ectopic papillae are innervated in the stabilizing beta-catenin mutant, whereas ectopic Dkk1 causes absence of lingual epithelial innervation. Thus, Wnt-beta-catenin signaling is critical for fungiform papilla and taste bud development. Altered regulation of this pathway may underlie evolutionary changes in taste papilla patterning.


Asunto(s)
Papilas Gustativas/embriología , Proteínas Wnt/fisiología , beta Catenina/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Ratones , Ratones Transgénicos , Morfogénesis/genética , Embarazo , Transducción de Señal/genética , Papilas Gustativas/crecimiento & desarrollo , beta Catenina/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...