Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 75
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Med Genet C Semin Med Genet ; : e32095, 2024 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-39022906

RESUMEN

Neurofibromatosis type 1 (NF-1) microdeletion syndrome accounts for 5 to 11% of individuals with NF-1. The aim of our study was to characterize a large cohort of individuals with NF-1 microdeletion syndrome and expand its natural history. We conducted a retrospective chart review from 1994 to 2024 of individuals with NF-1 microdeletion syndrome followed at two large Neurofibromatosis Clinics. This cohort consists of 57 individuals with NF-1 microdeletion syndrome (28 type-1, 4 type-2, 2 type-3, 9 atypical deletions, and 14 indeterminate). We note 38/56 (67.9%) with describable facial features, 25/57 (43.8%) with plexiform neurofibromas, and 3/57 (5.2%) with malignant peripheral nerve sheath tumors within the observed period. The most reported neurodevelopmental manifestations from school-age or older individuals included 39/49 (79.6%) with developmental delays, 35/49 (71.4%) with expressive and/or receptive speech delays, 33/41 (80.5%) with learning difficulties, and 23/42 (54.8%) with attention-deficit/hyperactivity disorder. Full-scale IQ testing data was available for 22 individuals (range: 50-96). Of the 21 adults in this cohort, 14/21 (66.7%) graduated from high school, and 4/21 (19.0%) had some college experience. Many individuals received academic support (i.e., special education, individual education plan). In this cohort, neurocognitive outcomes in adults varied more than typically reported in the literature.

2.
Genet Med ; 24(12): 2444-2452, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36107167

RESUMEN

PURPOSE: This study was undertaken to collect baseline growth parameters in children with achondroplasia who might enroll in interventional trials of vosoritide, and to establish a historical control. METHODS: In this prospective, observational study, participants (≤17 years) underwent a detailed medical history and physical examination and were followed every 3 months until they finished participating in the study by enrolling in an interventional trial or withdrawing. RESULTS: A total of 363 children were enrolled (28 centers, 8 countries). Mean (SD) follow up was 20.4 (15.0) months. In participants <1 year, mean annualized growth velocity (AGV) was 11.6 cm/year for girls and 14.6 cm/year for boys. By age 1 year, mean AGV decreased to 7.4 cm/year in girls and 7.1 cm/year in boys. By age 10 years, mean AGV decreased to 3.6 cm/year for both sexes. Mean height z-score in participants <1 year was -2.5 for girls and -3.2 for boys and decreased up to the age 5 years (-5.3 for girls; -4.6 for boys). Girls and boys had a disproportionate upper-to-lower body segment ratio. Mean ratio was highest in participants aged <1 year (2.9 for girls; 2.8 for boys) and decreased gradually to approximately 2 in both sexes from 4 years of age onward. CONCLUSION: This study represents one of the largest datasets of prospectively collected medical and longitudinal growth data in children with achondroplasia. It serves as a robust historical control to measure therapeutic interventions against and to further delineate the natural history of this condition.


Asunto(s)
Acondroplasia , Niño , Masculino , Femenino , Humanos , Preescolar , Estudios Prospectivos , Acondroplasia/epidemiología , Acondroplasia/genética , Acondroplasia/diagnóstico , Estatura
3.
Am J Med Genet A ; 188(1): 160-177, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34569146

RESUMEN

Robin sequence (RS), the triad of micrognathia, glossoptosis, and airway obstruction, is a major cause of respiratory distress and feeding difficulties in neonates. Robin sequence can be associated with other medical or developmental comorbidities in ~50% of cases ("syndromic" RS). As well, RS is variably associated with cleft palate (CP). Previous studies have not investigated differences in clinical characteristics of children with RS based on presence or absence of CP. We retrospectively reviewed 175 children with RS and compared genetic diagnoses, medical and developmental comorbidities, severity of airway obstruction, and feeding outcomes between those with and without CP. Strikingly, 45 of 45 (100%) children with RS without CP were classified as syndromic due to presence of comorbidities unrelated to RS, while 83 of 130 (64%) children with RS with CP were classified as syndromic. Among 128 children with syndromic RS, there were no differences in severity of airway obstruction, surgical intervention rate or type, or feeding outcome at 12 months based on CP status. Our findings support the conclusion that the pathogenesis of RS without CP is distinct from RS with CP and more likely to cause additional medical or developmental problems. Alternatively, children with RS without CP and without additional anomalies present may be under recognized.


Asunto(s)
Obstrucción de las Vías Aéreas , Fisura del Paladar , Micrognatismo , Síndrome de Pierre Robin , Obstrucción de las Vías Aéreas/diagnóstico , Obstrucción de las Vías Aéreas/genética , Niño , Fisura del Paladar/complicaciones , Fisura del Paladar/diagnóstico , Fisura del Paladar/genética , Humanos , Recién Nacido , Micrognatismo/complicaciones , Síndrome de Pierre Robin/diagnóstico , Síndrome de Pierre Robin/epidemiología , Síndrome de Pierre Robin/genética , Estudios Retrospectivos
4.
Lancet ; 396(10252): 684-692, 2020 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-32891212

RESUMEN

BACKGROUND: There are no effective therapies for achondroplasia. An open-label study suggested that vosoritide administration might increase growth velocity in children with achondroplasia. This phase 3 trial was designed to further assess these preliminary findings. METHODS: This randomised, double-blind, phase 3, placebo-controlled, multicentre trial compared once-daily subcutaneous administration of vosoritide with placebo in children with achondroplasia. The trial was done in hospitals at 24 sites in seven countries (Australia, Germany, Japan, Spain, Turkey, the USA, and the UK). Eligible patients had a clinical diagnosis of achondroplasia, were ambulatory, had participated for 6 months in a baseline growth study and were aged 5 to less than 18 years at enrolment. Randomisation was done by means of a voice or web-response system, stratified according to sex and Tanner stage. Participants, investigators, and trial sponsor were masked to group assignment. Participants received either vosoritide 15·0 µg/kg or placebo, as allocated, for the duration of the 52-week treatment period administered by daily subcutaneous injections in their homes by trained caregivers. The primary endpoint was change from baseline in mean annualised growth velocity at 52 weeks in treated patients as compared with controls. All randomly assigned patients were included in the efficacy analyses (n=121). All patients who received one dose of vosoritide or placebo (n=121) were included in the safety analyses. The trial is complete and is registered, with EudraCT, number, 2015-003836-11. FINDINGS: All participants were recruited from Dec 12, 2016, to Nov 7, 2018, with 60 assigned to receive vosoritide and 61 to receive placebo. Of 124 patients screened for eligibility, 121 patients were randomly assigned, and 119 patients completed the 52-week trial. The adjusted mean difference in annualised growth velocity between patients in the vosoritide group and placebo group was 1·57 cm/year in favour of vosoritide (95% CI [1·22-1·93]; two-sided p<0·0001). A total of 119 patients had at least one adverse event; vosoritide group, 59 (98%), and placebo group, 60 (98%). None of the serious adverse events were considered to be treatment related and no deaths occurred. INTERPRETATION: Vosoritide is an effective treatment to increase growth in children with achondroplasia. It is not known whether final adult height will be increased, or what the harms of long-term therapy might be. FUNDING: BioMarin Pharmaceutical.


Asunto(s)
Acondroplasia/tratamiento farmacológico , Péptido Natriurético Tipo-C/análogos & derivados , Osteogénesis , Absorciometría de Fotón , Acondroplasia/sangre , Adolescente , Biomarcadores/sangre , Estatura , Densidad Ósea , Niño , Preescolar , Colágeno Tipo X/sangre , Método Doble Ciego , Femenino , Humanos , Reacción en el Punto de Inyección , Inyecciones Subcutáneas , Masculino , Péptido Natriurético Tipo-C/uso terapéutico
5.
Genet Med ; 23(12): 2443-2447, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34341520

RESUMEN

PURPOSE: Achondroplasia is caused by pathogenic variants in the fibroblast growth factor receptor 3 gene that lead to impaired endochondral ossification. Vosoritide, an analog of C-type natriuretic peptide, stimulates endochondral bone growth and is in development for the treatment of achondroplasia. This phase 3 extension study was conducted to document the efficacy and safety of continuous, daily vosoritide treatment in children with achondroplasia, and the two-year results are reported. METHODS: After completing at least six months of a baseline observational growth study, and 52 weeks in a double-blind, placebo-controlled study, participants were eligible to continue treatment in an open-label extension study, where all participants received vosoritide at a dose of 15.0 µg/kg/day. RESULTS: In children randomized to vosoritide, annualized growth velocity increased from 4.26 cm/year at baseline to 5.39 cm/year at 52 weeks and 5.52 cm/year at week 104. In children who crossed over from placebo to vosoritide in the extension study, annualized growth velocity increased from 3.81 cm/year at week 52 to 5.43 cm/year at week 104. No new adverse effects of vosoritide were detected. CONCLUSION: Vosoritide treatment has safe and persistent growth-promoting effects in children with achondroplasia treated daily for two years.


Asunto(s)
Acondroplasia , Péptido Natriurético Tipo-C , Acondroplasia/tratamiento farmacológico , Acondroplasia/genética , Niño , Método Doble Ciego , Humanos , Péptido Natriurético Tipo-C/análogos & derivados , Péptido Natriurético Tipo-C/uso terapéutico , Resultado del Tratamiento
6.
Am J Med Genet A ; 185(2): 413-423, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33247512

RESUMEN

Mandibulofacial dysostosis with microcephaly (MFDM) is due to haploinsufficiency of spliceosomal GTPase EFTUD2. Features include microcephaly, craniofacial dysmorphology, developmental disability, and other anomalies. We surveyed parents of individuals with MFDM to expand knowledge about health, development, and parental concerns. Participants included attendees of the inaugural MFDM family conference in June 2019 and members of the MFDM online group. To explore MFDM variable expressivity, we offered targeted Sanger sequencing for untested parents. Forty-seven parents participated in the survey. 59% of individuals with MFDM were male, with mean age 6.4 years (range 8 months to 49 years). Similar to the literature (n = 123), common features include microcephaly, cleft palate, choanal stenosis, tracheoesophageal fistula, heart problems, and seizures. New information includes airway intervention details, age-based developmental outcomes, rate of vision refractive errors, and lower incidences of prematurity and IUGR. Family concerns focused on development, communication, and increased support. Targeted Sanger sequencing for families of seven individuals demonstrated de novo variants, for a total of 91.9% de novo EFTUD2 variants (n = 34/37). This study reports the largest single cohort of individuals with MFDM, expands phenotypic spectrum and inheritance patterns, improves understanding of developmental outcomes and care needs, and identifies development as the biggest concern for parents.


Asunto(s)
Disostosis Mandibulofacial/genética , Microcefalia/genética , Factores de Elongación de Péptidos/genética , Ribonucleoproteína Nuclear Pequeña U5/genética , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Femenino , GTP Fosfohidrolasas/genética , Predisposición Genética a la Enfermedad , Haploinsuficiencia/genética , Humanos , Lactante , Masculino , Disostosis Mandibulofacial/complicaciones , Disostosis Mandibulofacial/patología , Microcefalia/complicaciones , Microcefalia/patología , Persona de Mediana Edad , Mutación/genética , Fenotipo , Empalmosomas/genética , Empalmosomas/patología , Adulto Joven
7.
Am J Med Genet C Semin Med Genet ; 184(3): 644-655, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32888375

RESUMEN

Mosaic genetic mutations may be somatic, germline, or "gonosomal" and have the potential to cause genetic syndromes, disorders, or malformations. Mutations can occur at any point in embryonic development and the timing determines the extent of distribution of the mutation throughout the body and different tissue types. The eye and visual pathway offer a unique opportunity to study somatic and gonosomal mosaic mutations as the eye consists of tissues derived from all three germ layers allowing disease pathology to be assessed with noninvasive imaging. In this review, we describe systemic and ocular manifestations in a child with mosaic Coffin-Siris syndrome. The patient presented with a significant medical history of accommodative esotropia and hyperopia, macrocephaly, polydactyly, global developmental delay, hypotonia, ureteropelvic junction (UPJ) obstruction, and brain MRI abnormalities. The ophthalmic findings in this patient were nonspecific, however, they are consistent with ocular manifestations reported in other patients with Coffin-Siris syndrome. We also review ophthalmic findings of select mosaic chromosomal and single-gene disorders. Ophthalmic assessment alongside clinical genetic testing may play an important role in diagnosis of genetic syndromes as well as understanding disease pathology, particularly when mosaicism plays a role.


Asunto(s)
Anomalías Múltiples/genética , Encéfalo/diagnóstico por imagen , Proteínas de Unión al ADN/genética , Cara/anomalías , Predisposición Genética a la Enfermedad , Deformidades Congénitas de la Mano/genética , Discapacidad Intelectual/genética , Micrognatismo/genética , Cuello/anomalías , Factores de Transcripción/genética , Anomalías Múltiples/diagnóstico por imagen , Anomalías Múltiples/patología , Encéfalo/anomalías , Niño , Preescolar , Cara/diagnóstico por imagen , Cara/patología , Femenino , Deformidades Congénitas de la Mano/diagnóstico por imagen , Deformidades Congénitas de la Mano/patología , Humanos , Lactante , Discapacidad Intelectual/diagnóstico por imagen , Discapacidad Intelectual/patología , Masculino , Micrognatismo/diagnóstico por imagen , Micrognatismo/patología , Mosaicismo , Mutación/genética , Cuello/diagnóstico por imagen , Cuello/patología , Proteínas Nucleares/genética , Fenotipo
8.
Genesis ; 57(1): e23259, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30375152

RESUMEN

A recent convergence of technological innovations has re-energized the ability to apply genetics to research in human craniofacial development. Next-generation exome and whole genome sequencing have significantly dropped in price, making it relatively trivial to sequence and analyze patients and families with congenital craniofacial anomalies. A concurrent revolution in genome editing with the use of the CRISPR-Cas9 system enables the rapid generation of animal models, including mouse, which can precisely recapitulate human variants. Here, we summarize the choices currently available to the research community. We illustrate this approach with the study of a family with a novel craniofacial syndrome with dominant inheritance pattern. The genomic analysis suggested a causal variant in AMOTL1 which we modeled in mice. We also made a novel deletion allele of Amotl1. Our results indicate that Amotl1 is not required in the mouse for survival to weaning. Mice carrying the variant identified in the human sequencing studies, however, do not survive to weaning in normal ratios. The cause of death is not understood for these mice complicating our conclusions about the pathogenicity in the index patient. Thus, we highlight some of the powerful opportunities and confounding factors confronting current craniofacial genetic research.


Asunto(s)
Anomalías Craneofaciales/genética , Modelos Animales de Enfermedad , Proteínas de la Membrana/genética , Adulto , Angiomotinas , Proteína 1 Similar a la Angiopoyetina , Animales , Anomalías Craneofaciales/patología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Análisis de Secuencia de ADN/métodos
9.
Am J Hum Genet ; 96(5): 765-74, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25913037

RESUMEN

We report three individuals with a cranioskeletal malformation syndrome that we define as acrofacial dysostosis, Cincinnati type. Each individual has a heterozygous mutation in POLR1A, which encodes a core component of RNA polymerase 1. All three individuals exhibit varying degrees of mandibulofacial dysostosis, and two additionally have limb anomalies. Consistent with this observation, we discovered that polr1a mutant zebrafish exhibited cranioskeletal anomalies mimicking the human phenotype. polr1a loss of function led to perturbed ribosome biogenesis and p53-dependent cell death, resulting in a deficiency of neural-crest-derived skeletal precursor cells and consequently craniofacial anomalies. Our findings expand the genotypic and phenotypic heterogeneity of congenital acrofacial disorders caused by disruption of ribosome biogenesis.


Asunto(s)
Deformidades Congénitas de las Extremidades/genética , Disostosis Mandibulofacial/genética , ARN Polimerasa I/genética , Ribosomas/genética , Animales , Muerte Celular/genética , Genotipo , Humanos , Deformidades Congénitas de las Extremidades/fisiopatología , Disostosis Mandibulofacial/fisiopatología , Mutación , Cresta Neural/crecimiento & desarrollo , Cresta Neural/patología , Ribosomas/patología , Pez Cebra
10.
Am J Hum Genet ; 97(1): 6-21, 2015 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-26140447

RESUMEN

In 1995, the American Society of Human Genetics (ASHG) and American College of Medical Genetics and Genomics (ACMG) jointly published a statement on genetic testing in children and adolescents. In the past 20 years, much has changed in the field of genetics, including the development of powerful new technologies, new data from genetic research on children and adolescents, and substantial clinical experience. This statement represents current opinion by the ASHG on the ethical, legal, and social issues concerning genetic testing in children. These recommendations are relevant to families, clinicians, and investigators. After a brief review of the 1995 statement and major changes in genetic technologies in recent years, this statement offers points to consider on a broad range of test technologies and their applications in clinical medicine and research. Recommendations are also made for record and communication issues in this domain and for professional education.


Asunto(s)
Pruebas Genéticas/ética , Pruebas Genéticas/legislación & jurisprudencia , Pruebas Genéticas/tendencias , Genética/historia , Genómica/métodos , Consentimiento Informado de Menores/psicología , Adolescente , Niño , Tamización de Portadores Genéticos , Genómica/ética , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Recién Nacido , Análisis por Micromatrices/métodos , Análisis por Micromatrices/tendencias , Farmacogenética/métodos
11.
Am J Hum Genet ; 96(4): 519-31, 2015 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-25772936

RESUMEN

The endothelin receptor type A (EDNRA) signaling pathway is essential for the establishment of mandibular identity during development of the first pharyngeal arch. We report four unrelated individuals with the syndrome mandibulofacial dysostosis with alopecia (MFDA) who have de novo missense variants in EDNRA. Three of the four individuals have the same substitution, p.Tyr129Phe. Tyr129 is known to determine the selective affinity of EDNRA for endothelin 1 (EDN1), its major physiological ligand, and the p.Tyr129Phe variant increases the affinity of the receptor for EDN3, its non-preferred ligand, by two orders of magnitude. The fourth individual has a somatic mosaic substitution, p.Glu303Lys, and was previously described as having Johnson-McMillin syndrome. The zygomatic arch of individuals with MFDA resembles that of mice in which EDNRA is ectopically activated in the maxillary prominence, resulting in a maxillary to mandibular transformation, suggesting that the p.Tyr129Phe variant causes an EDNRA gain of function in the developing upper jaw. Our in vitro and in vivo assays suggested complex, context-dependent effects of the EDNRA variants on downstream signaling. Our findings highlight the importance of finely tuned regulation of EDNRA signaling during human craniofacial development and suggest that modification of endothelin receptor-ligand specificity was a key step in the evolution of vertebrate jaws.


Asunto(s)
Alopecia/genética , Disostosis Mandibulofacial/genética , Receptor de Endotelina A/genética , Alopecia/patología , Animales , Secuencia de Bases , Endotelina-1/metabolismo , Exoma/genética , Humanos , Hibridación in Situ , Disostosis Mandibulofacial/patología , Datos de Secuencia Molecular , Morfolinos/genética , Mutación Missense/genética , Linaje , ARN Mensajero/administración & dosificación , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor de Endotelina A/metabolismo , Análisis de Secuencia de ADN , Síndrome , Tomografía Computarizada por Rayos X , Pez Cebra , Cigoma/patología
12.
Pediatr Radiol ; 48(7): 1032-1034, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29541808

RESUMEN

Fetal ventriculomegaly is a common referral for prenatal MRI, with possible etiologies being hydrocephalus and hydranencephaly. The underlying cause of hydranencephaly is unknown, but many have suggested that the characteristic supratentorial injury is related to idiopathic bilateral occlusions of the internal carotid arteries from an acquired or destructive event. Fowler syndrome is a rare genetic disorder that causes fetal akinesia and a proliferative vasculopathy that can result in an apparent hydranencephaly-hydrocephaly complex. On prenatal imaging, the presence of significant parenchymal loss in the supratentorial and infratentorial brain is a clue to the diagnosis, which should prompt early genetic testing.


Asunto(s)
Anomalías Múltiples/diagnóstico por imagen , Enfermedades Fetales/diagnóstico por imagen , Hidranencefalia/diagnóstico por imagen , Hidrocefalia/diagnóstico por imagen , Imagen por Resonancia Magnética/métodos , Diagnóstico Prenatal/métodos , Anomalías Múltiples/genética , Aborto Inducido , Adulto , Diagnóstico Diferencial , Femenino , Enfermedades Fetales/genética , Pruebas Genéticas , Humanos , Hidranencefalia/genética , Hidrocefalia/genética , Mutación , Embarazo , Síndrome
13.
Hum Mol Genet ; 24(12): 3399-409, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25759469

RESUMEN

Autosomal dominant omodysplasia is a rare skeletal dysplasia characterized by short humeri, radial head dislocation, short first metacarpals, facial dysmorphism and genitourinary anomalies. We performed next-generation whole-exome sequencing and comparative analysis of a proband with omodysplasia, her unaffected parents and her affected daughter. We identified a de novo mutation in FRIZZLED2 (FZD2) in the proband and her daughter that was not found in unaffected family members. The FZD2 mutation (c.1644G>A) changes a tryptophan residue at amino acid 548 to a premature stop (p.Trp548*). This altered protein is still produced in vitro, but we show reduced ability of this mutant form of FZD2 to interact with its downstream target DISHEVELLED. Furthermore, expressing the mutant form of FZD2 in vitro is not able to facilitate the cellular response to canonical Wnt signaling like wild-type FZD2. We therefore conclude that the FRIZZLED2 mutation is a de novo, novel cause for autosomal dominant omodysplasia.


Asunto(s)
Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Húmero/anomalías , Huesos del Metacarpo/anomalías , Mutación , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Vía de Señalización Wnt , Adulto , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Huesos/diagnóstico por imagen , Huesos/patología , Análisis Mutacional de ADN , Exoma , Facies , Femenino , Receptores Frizzled/química , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Húmero/metabolismo , Lactante , Huesos del Metacarpo/metabolismo , Osteocondrodisplasias/diagnóstico , Linaje , Fenotipo , Unión Proteica , Transporte de Proteínas , Radiografía
14.
Mol Genet Metab ; 122(1-2): 4-17, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28888853

RESUMEN

Hypophosphatasia (HPP) is a rare, inherited, systemic, metabolic disorder caused by autosomal recessive mutations or a single dominant-negative mutation in the gene encoding tissue-nonspecific alkaline phosphatase (TNSALP). The disease is associated with a broad range of signs, symptoms, and complications, including impaired skeletal mineralization, altered calcium and phosphate metabolism, recurrent fractures, pain, respiratory problems, impaired growth and mobility, premature tooth loss, developmental delay, and seizures. Asfotase alfa is a human, recombinant enzyme replacement therapy that is approved in many countries for the treatment of patients with HPP. To address the unmet need for guidance in the monitoring of patients receiving asfotase alfa, an international panel of physicians with experience in diagnosing and managing HPP convened in May 2016 to discuss treatment monitoring parameters. The panel discussions focused on recommendations for assessing and monitoring patients after the decision to treat with asfotase alfa had been made and did not include recommendations for whom to treat. Based on the consensus of panel members, this review provides guidance on the monitoring of patients with HPP during treatment with asfotase alfa, including recommendations for laboratory, efficacy, and safety assessments and the frequency with which these should be performed during the course of treatment. Recommended assessments are based on patient age and include regular monitoring of biochemistry, skeletal radiographs, respiratory function, growth, pain, mobility and motor function, and quality of life. Because of the systemic presentation of HPP, a coordinated, multidisciplinary, team-based, patient-focused approach is recommended in the management of patients receiving asfotase alfa. Monitoring of efficacy and safety outcomes must be tailored to the individual patient, depending on medical history, clinical manifestations, availability of resources in the clinical setting, and the clinician's professional judgment.


Asunto(s)
Fosfatasa Alcalina/uso terapéutico , Enfermedades Óseas Metabólicas/tratamiento farmacológico , Monitoreo de Drogas/métodos , Hipofosfatasia/tratamiento farmacológico , Inmunoglobulina G/uso terapéutico , Proteínas Recombinantes de Fusión/uso terapéutico , Fosfatasa Alcalina/metabolismo , Enfermedades Óseas Metabólicas/fisiopatología , Terapia de Reemplazo Enzimático , Humanos , Hipofosfatasia/diagnóstico por imagen , Hipofosfatasia/fisiopatología
15.
J Allergy Clin Immunol ; 137(1): 179-187.e10, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26194542

RESUMEN

BACKGROUND: Kabuki syndrome (KS) is a complex multisystem developmental disorder associated with mutation of genes encoding histone-modifying proteins. In addition to craniofacial, intellectual, and cardiac defects, KS is also characterized by humoral immune deficiency and autoimmune disease, yet no detailed molecular characterization of the KS-associated immune phenotype has been reported. OBJECTIVE: We sought to characterize the humoral immune defects found in patients with KS with lysine methyltransferase 2D (KMT2D) mutations. METHODS: We comprehensively characterized B-cell function in a cohort (n = 13) of patients with KS (age, 4 months to 27 years). RESULTS: Three quarters (77%) of the cohort had a detectable heterozygous KMT2D mutation (50% nonsense, 20% splice site, and 30% missense mutations), and 70% of the reported mutations are novel. Among the patients with KMT2D mutations (KMT2D(Mut/+)), hypogammaglobulinemia was detected in all but 1 patient, with IgA deficiency affecting 90% of patients and a deficiency in at least 1 other isoform seen in 40% of patients. Numbers of total memory (CD27(+)) and class-switched memory B cells (IgM(-)) were significantly reduced in patients with KMT2D(Mut/+) mutations compared with numbers in control subjects (P < .001). Patients with KMT2D(Mut/+) mutations also had significantly reduced rates of somatic hypermutation in IgG (P = .003) but not IgA or IgM heavy chain sequences. Impaired terminal differentiation was noted in primary B cells from patients with KMT2D(Mut/+) mutations. Autoimmune pathology was observed in patients with missense mutations affecting the SET domain and its adjacent domains. CONCLUSIONS: In patients with KS, autosomal dominant KMT2D mutations are associated with dysregulation of terminal B-cell differentiation, leading to humoral immune deficiency and, in some cases, autoimmunity. All patients with KS should undergo serial clinical immune evaluations.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Múltiples/inmunología , Linfocitos B/citología , Proteínas de Unión al ADN/genética , Cara/anomalías , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/inmunología , Proteínas de Neoplasias/genética , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/inmunología , Adolescente , Adulto , Agammaglobulinemia/genética , Agammaglobulinemia/inmunología , Linfocitos B/inmunología , Diferenciación Celular , Niño , Preescolar , Humanos , Lactante , Mutación , Adulto Joven
16.
BMC Med Genet ; 17(1): 86, 2016 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-27871226

RESUMEN

BACKGROUND: A new disease class of syndromes, described as linkeropathies, which are derived from defects in the glycosaminoglycan-linker region as well as glycosaminoglycan-side chains of proteoglycans is increasingly being recognized as a cause of human disease. Proteoglycans are an essential component of the extracellular matrix. Defects in the enzymatic process of proteoglycan synthesis broadly occur due to the incorrect addition of side chains. Previously, homozygous missense variants within the B3GAT3 gene encoding beta 1,3 glucuronyltransferase 3(GlcAT-I) responsible for the biosynthesis of glycosaminoglycans have been described in 7 individuals. CASE PRESENTATION: In this study, a 4-year-old patient with a severe phenotype of osteoporosis, hypotonia, joint laxity, fractures, scoliosis, biscuspid aortic valve and myopia was referred for next generation sequencing after extensive negative clinical testing. Whole exome sequencing was performed on the proband and his unaffected parents to identify the molecular basis of his disease. Sequencing revealed compound heterozygous variants in B3GAT3: c.1A > G (p.Met1?) and c.671 T > A (p.L224Q). Clinical and in vitro functional studies were then completed to verify the pathogenicity of the genotype and further characterize the functional basis of the patient's disease demonstrating the patient had a decrease both in the protein level of B3GAT3 and in the glucuronyltransferase activity when compared to control samples. Independent in vitro assessment of each variant confirmed the B3GAT3: c.1A > G (p.Met1?) variant is functionally null and the c.671 T > A (p.L224Q) missense variant has significantly reduced glucuronyltransferase activity (~3% of control). CONCLUSIONS: This is the first report of a patient with compound heterozygosity for a null variant in trans with a missense in B3GAT3 resulting in a severe phenotype, expanding both the genotypic and phenotypic spectrum of B3GAT3-related disease.


Asunto(s)
Enfermedades Óseas Metabólicas/genética , Fracturas Óseas/genética , Variación Genética , Glucuronosiltransferasa/genética , Enfermedades Óseas Metabólicas/patología , Preescolar , ADN/química , ADN/aislamiento & purificación , ADN/metabolismo , Genotipo , Glucuronosiltransferasa/metabolismo , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Mutación Missense , Linaje , Fenotipo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética
17.
J Pediatr ; 173: 143-8, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26987801

RESUMEN

OBJECTIVES: To assess whether children with Down syndrome in the US are at an increased risk for obesity, we determined the obesity prevalence and analyzed obesity development throughout childhood in a cohort of children with Down syndrome. In addition, we analyzed a comorbidity that is associated with Down syndrome and obesity, obstructive sleep apnea syndrome (OSAS). STUDY DESIGN: This study was a retrospective chart review that evaluated 303 children ages 2 through 18 years with a diagnosis of Down syndrome. All children were patients at Cincinnati Children's Hospital Medical Center with multiple height and weight measurements. To determine obesity burden, the rate of obesity was compared with a local control cohort using contingency tables. Change in obesity rate through time was determined with mixed models. Association of obesity with OSAS was determined with contingency tables. RESULTS: We evaluated 303 individuals, 47.8% of whom were obese (body mass index ≥95th percentile for age and sex). This was significantly higher than the general pediatric population, which had a 12.1% obesity rate (P < .0001). Body mass index z-scores did not change markedly over time (P = .40). The majority of children with Down syndrome also had OSAS (74.0% of the 177 children who had polysomnography studies). However, OSAS risk was elevated in obese children (risk ratio = 2.4, P = .0015). CONCLUSIONS: Our results indicate that children with Down syndrome are at a substantial risk for obesity and OSAS. These findings support the need for more aggressive weight management in early childhood and throughout the lifespan.


Asunto(s)
Síndrome de Down/complicaciones , Obesidad Infantil/complicaciones , Adolescente , Estudios de Casos y Controles , Niño , Preescolar , Femenino , Humanos , Masculino , Polisomnografía , Prevalencia , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Apnea Obstructiva del Sueño/complicaciones
18.
Am J Med Genet A ; 170A(2): 487-491, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26581443

RESUMEN

The frontonasal dysplasias are a group of craniofacial phenotypes characterized by hypertelorism, nasal clefting, frontal bossing, and abnormal hairline. These conditions are caused by recessive mutations in members of the aristaless gene family, resulting in abnormal cranial neural crest migration and differentiation. We report a family with a dominantly inherited craniofacial phenotype comprised of frontal bossing with high hairline, ptosis, hypertelorism, broad nasal tip, large anterior fontanelle, cranial base anomalies, and sagittal synostosis. Chromosomal microarray identified a heterozygous 108.3 kilobase deletion of chromosome 2p21 segregating with phenotype and limited to the sine oculis homeobox gene SIX2 and surrounding noncoding DNA. Similar to the human SIX2 deletion phenotype, one mouse model of frontonasal dysplasia, brachyrrhine, exhibits dominant inheritance and impaired cranial base chondrogenesis associated with reduced Six2 expression. We report the first human autosomal dominant frontonasal dysplasia syndrome associated with SIX2 deletion and with phenotypic similarities to murine models of Six2 Loss-of-function.


Asunto(s)
Anomalías Craneofaciales/genética , Cara/anomalías , Eliminación de Gen , Proteínas de Homeodominio/genética , Mutación/genética , Proteínas del Tejido Nervioso/genética , Adulto , Animales , Anomalías Craneofaciales/patología , Cara/patología , Femenino , Heterocigoto , Humanos , Lactante , Ratones , Fenotipo , Síndrome
19.
J Pediatr ; 167(6): 1404-8.e1, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26477869

RESUMEN

OBJECTIVE: To examine the decisions of pediatric primary care physicians about their diagnostic evaluation for a child with suspected global developmental delay (GDD). STUDY DESIGN: A survey was mailed to a sample of pediatricians (n = 600) and family physicians (n = 600) randomly selected from the American Medical Association Physician Masterfile. The survey contained a clinical vignette describing a 9-month-old nondysmorphic boy with GDD. Participants were asked their initial evaluation steps (test, refer, or both test and refer) and what types of referral and/or testing they would pursue. We examined bivariate associations between physician/clinical practice characteristics and participants' evaluation decision. RESULTS: More pediatricians than family physicians completed the survey (response rates: 55% vs 38%). Almost three-quarters of the respondents (74%) reported that their first step in a diagnostic evaluation would be to refer the child without testing, 22% would test only, and 4% would both test and refer. As their initial step, most physicians referred to a developmental pediatrician (58%), and only 5% would refer to a geneticist. The most commonly ordered test was general biochemical testing (64%). The most commonly ordered genetic test was a karyotype (39%). CONCLUSIONS: When evaluating a child with GDD, few primary care physicians would order genetic testing or refer to a genetics specialist as a first evaluation step. Future studies should examine both barriers to and utilization of a genetic evaluation for children with GDD.


Asunto(s)
Discapacidades del Desarrollo/diagnóstico , Diagnóstico por Imagen , Pruebas Genéticas , Médicos de Familia/normas , Pautas de la Práctica en Medicina , Atención Primaria de Salud/métodos , Femenino , Humanos , Lactante , Masculino , Persona de Mediana Edad , Estados Unidos
20.
J Pediatr ; 167(4): 851-856.e1, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26233602

RESUMEN

OBJECTIVE: To evaluate the utility of screening brain/orbital magnetic resonance imaging (MRI) in a large population of children with neurofibromatosis type 1 (NF1) over a 20-year period. STUDY DESIGN: A retrospective analysis of clinical and imaging data from children with NF1 seen at a single center between 1990 and 2010 was performed. RESULTS: During the 20-year study period, 826 individuals with NF1 (402 females, 424 males) ages 1-9 years were screened for optic pathway gliomas (OPGs) using brain/orbital MRI; 18% were identified with OPGs with a median age at detection of 3 years. Fifteen percent of patients with OPGs had radiologic or clinical progression requiring therapy. Children with chiasmatic and postchiasmatic tumors were more likely to require therapy compared with patients with prechiasmatic OPGs (P < .0001). Patients with visual deficits at the time of diagnosis were more likely to experience visual decline despite therapy when compared with patients treated based on radiologic progression (P < .012). CONCLUSIONS: Our findings confirm that chiasmatic and postchiasmatic OPG in children with NF1 have the highest risk for progression and vision loss. Early identification of OPG by screening MRI prior to the development of vision loss may lead to improved visual outcomes. Children with negative brain and orbital MRI screening at age 15 months or later did not develop symptomatic OPGs.


Asunto(s)
Imagen por Resonancia Magnética , Neurofibromatosis 1/diagnóstico , Glioma del Nervio Óptico/diagnóstico , Neoplasias del Nervio Óptico/diagnóstico , Encéfalo/patología , Niño , Preescolar , Comorbilidad , Progresión de la Enfermedad , Femenino , Humanos , Lactante , Masculino , Neurofibromatosis 1/complicaciones , Glioma del Nervio Óptico/complicaciones , Neoplasias del Nervio Óptico/complicaciones , Estudios Retrospectivos , Trastornos de la Visión/complicaciones , Trastornos de la Visión/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA