Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Prenat Diagn ; 44(2): 237-246, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37632214

RESUMEN

OBJECTIVE: Recurrent deletions involving 17q12 are associated with a variety of clinical phenotypes, including congenital abnormalities of the kidney and urinary tract (CAKUT), maturity onset diabetes of the young, type 5, and neurodevelopmental disorders. Structural and/or functional renal disease is the most common phenotypic feature, although the prenatal renal phenotypes and the postnatal correlates have not been well characterized. METHOD: We reviewed pre- and postnatal medical records of 26 cases with prenatally or postnatally identified 17q12/HNF1B microdeletions (by chromosomal microarray or targeted gene sequencing), obtained through a multicenter collaboration. We specifically evaluated 17 of these cases (65%) with reported prenatal renal ultrasound findings. RESULTS: Heterogeneous prenatal renal phenotypes were noted, most commonly renal cysts (41%, n = 7/17) and echogenic kidneys (41%), although nonspecific dysplasia, enlarged kidneys, hydronephrosis, pelvic kidney with hydroureter, and lower urinary tract obstruction were also reported. Postnatally, most individuals developed renal cysts (73%, 11/15 live births), and there were no cases of end-stage renal disease during childhood or the follow-up period. CONCLUSION: Our findings demonstrate that copy number variant analysis to assess for 17q12 microdeletion should be considered for a variety of prenatally detected renal anomalies. It is important to distinguish 17q12 microdeletion from other etiologies of CAKUT as the prognosis for renal function and presence of associated findings are distinct and may influence pregnancy and postnatal management.


Asunto(s)
Enfermedades Renales Quísticas , Enfermedades Renales , Anomalías Urogenitales , Reflujo Vesicoureteral , Embarazo , Femenino , Humanos , Deleción Cromosómica , Riñón/diagnóstico por imagen , Riñón/anomalías , Enfermedades Renales/congénito , Fenotipo , Enfermedades Renales Quísticas/diagnóstico por imagen , Enfermedades Renales Quísticas/genética , Factor Nuclear 1-beta del Hepatocito/genética , Estudios Multicéntricos como Asunto
2.
Am J Hum Genet ; 106(6): 872-884, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32470376

RESUMEN

Genome-wide analysis methods, such as array comparative genomic hybridization (CGH) and whole-genome sequencing (WGS), have greatly advanced the identification of structural variants (SVs) in the human genome. However, even with standard high-throughput sequencing techniques, complex rearrangements with multiple breakpoints are often difficult to resolve, and predicting their effects on gene expression and phenotype remains a challenge. Here, we address these problems by using high-throughput chromosome conformation capture (Hi-C) generated from cultured cells of nine individuals with developmental disorders (DDs). Three individuals had previously been identified as harboring duplications at the SOX9 locus and six had been identified with translocations. Hi-C resolved the positions of the duplications and was instructive in interpreting their distinct pathogenic effects, including the formation of new topologically associating domains (neo-TADs). Hi-C was very sensitive in detecting translocations, and it revealed previously unrecognized complex rearrangements at the breakpoints. In several cases, we observed the formation of fused-TADs promoting ectopic enhancer-promoter interactions that were likely to be involved in the disease pathology. In summary, we show that Hi-C is a sensible method for the detection of complex SVs in a clinical setting. The results help interpret the possible pathogenic effects of the SVs in individuals with DDs.


Asunto(s)
Cromosomas Humanos/genética , Discapacidades del Desarrollo/genética , Genoma Humano/genética , Conformación Molecular , Translocación Genética/genética , Ensamble y Desensamble de Cromatina/genética , Puntos de Rotura del Cromosoma , Estudios de Cohortes , Humanos , Factor de Transcripción SOX9/genética , Duplicaciones Segmentarias en el Genoma/genética
3.
Genet Med ; 24(10): 2167-2179, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36040446

RESUMEN

PURPOSE: The field of genetics and genomics continues to expand at an unprecedented pace. As scientific knowledge is translated to clinical practice, genomic information is routinely being used in preventive, diagnostic, and therapeutic decision-making across a variety of clinical practice areas. As adoption of genomic medicine further evolves, health professionals will be required to stay abreast of new genetic discoveries and technologies and implementation of these advances within their scope of practice will be indicated. METHODS: The Association of Professors of Human and Medical Genetics previously developed medical school genetics core competencies, last updated in 2013. The competencies were reviewed and updated through a structured approach incorporating a modified Delphi method. RESULTS: The updated Association of Professors of Human and Medical Genetics core competencies are presented. Current revisions include competencies that are concise, specific, and assessable. In addition, they incorporate recent advances in clinical practice and promote equity and inclusion in clinical care. CONCLUSION: The 2022 competencies will serve as a guide for medical school leadership and educators involved in curriculum development, implementation, and assessment. Use of these competencies across the undergraduate medical curricula will foster knowledge, skills, and behaviors required in medical practice across a wide range of specialties.


Asunto(s)
Educación de Pregrado en Medicina , Genética Médica , Competencia Clínica , Consenso , Curriculum , Genética Médica/educación , Genómica/educación , Humanos
4.
Hum Genet ; 140(4): 681-690, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33389145

RESUMEN

PURPOSE: Nuclear receptor binding SET domain protein 1, NSD1, encodes a histone methyltransferase H3K36. NSD1 is responsible for the phenotype of the reciprocal 5q35.2q35.3 microdeletion-microduplication syndromes. We expand the phenotype and demonstrate the functional role of NSD1 in microduplication 5q35 syndrome. METHODS: Through an international collaboration, we report nine new patients, contributing to the emerging phenotype, highlighting psychiatric phenotypes in older affected individuals. Focusing specifically on the undergrowth phenotype, we have modeled the effects of Mes-4/NSD overexpression in Drosophila melanogaster. RESULTS: The individuals (including a family) from diverse backgrounds with duplications ranging in size from 0.6 to 4.5 Mb, have a consistent undergrowth phenotype. Mes-4 overexpression in the developing wing causes undergrowth, increased H3K36 methylation, and increased apoptosis. We demonstrate that altering the levels of insulin receptor (IR) rescues the apoptosis and the wing undergrowth phenotype, suggesting changes in mTOR pathway signaling. Leucine supplementation rescued Mes-4/NSD induced cell death, demonstrating decreased mTOR signaling caused by NSD1. CONCLUSION: Given that we show mTOR inhibition as a likely mechanism and amelioration of the phenotype by leucine supplementation in a fly model, we suggest further studies should evaluate the therapeutic potential of leucine or branched chain amino acids as an adjunct possible treatment to ameliorate human growth and psychiatric phenotypes and propose inclusion of 5q35-microduplication as part of the differential diagnosis for children and adults with delayed bone age, short stature, microcephaly, developmental delay, and psychiatric phenotypes.


Asunto(s)
Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 5 , Duplicación de Gen , N-Metiltransferasa de Histona-Lisina/genética , Serina-Treonina Quinasas TOR/metabolismo , Adolescente , Adulto , Animales , Caspasas/metabolismo , Muerte Celular , Niño , Preescolar , Regulación hacia Abajo , Drosophila melanogaster , Femenino , Humanos , Leucina/metabolismo , Leucina/farmacología , Masculino , Linaje , Fenotipo , Transducción de Señal , Adulto Joven
5.
Am J Med Genet A ; 185(3): 894-900, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33369065

RESUMEN

Neuroligin 4 X-linked (NLGN4X) is an X-linked postsynaptic scaffolding protein, with functional role in excitatory synapsis development and maintenance, that has been associated with neuropsychiatric disorders such as intellectual disability, autism spectrum disorders (ASD), anxiety, attention deficit hyperactivity disorder (ADHD), and Tourette's syndrome. Chromosomal microarray analysis identified a paternally inherited, 445 Kb deletion on Xp22.3 that includes the entire NLGN4X in a 2.5 year old female (46,XX) with congenital hypotonia, strabismus, ASD, and increased aggressive behavioral issues. Her family history is significant for a mother with learning disabilities, a father with anxiety, major depressive disorder, and substance abuse, as well as two maternal half-brothers with developmental delays. X-inactivation studies in the proband's blood showed random X-inactivation despite the presence of an abnormal X chromosome. Furthermore, trio exome sequencing did not reveal any other deleterious variant that could explain her phenotype. Our report describes the first example of a paternally inherited NLGN4X microdeletion as the genetic etiology of ASD in a female proband, and the psychiatric phenotypes in the father. It also provides further evidence that NLGN4X is sensitive to dosage changes in females, and can contribute to a variety of psychiatric features within the same family.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Cromosomas Humanos X/genética , Discapacidades del Desarrollo/genética , Eliminación de Gen , Genes Ligados a X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Síndrome de Tourette/genética , Adulto , Agresión , Ansiedad/genética , Trastorno del Espectro Autista/genética , Trastornos de la Conducta Infantil/genética , Preescolar , Femenino , Humanos , Hibridación Fluorescente in Situ , Discapacidades para el Aprendizaje/genética , Masculino , Anamnesis , Análisis por Micromatrices , Herencia Paterna , Convulsiones/genética , Estrabismo/genética , Trastornos Relacionados con Sustancias/genética , Secuenciación del Exoma , Inactivación del Cromosoma X
6.
Am J Med Genet A ; 185(10): 3042-3047, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34196458

RESUMEN

22q11.2 deletion syndrome (22q11.2 DS, MIM #188400) is the most common chromosomal microdeletion with an incidence of 1 in 4000 live births. 22q11.2 DS patients present with varying penetrance and a broad phenotypic spectrum including dysmorphic features, congenital heart defects, hypoplastic thymus and T-cell deficiency, and hypocalcemia. The typical deletion spans 3 Mb between 4 large blocks of repetitive DNA, known as low copy repeats (LCRs), on chromosome 22 (LCR22) A and D. This deletion is found in ~85% of 22q11.2 DS patients, while only 4-5% have central LCR22B-D (1.5 Mb) and LCR22C-D (0.7 Mb) deletions. We report on a prenatally diagnosed, inherited case of central LCR22B-D 22q11.2 DS, born to a 22-year-old female with multiple autoimmune disorders. These include Sjogren's-syndrome-related antigen A (SSA+) severe systemic lupus erythematosus (SLE) with cutaneous and discoid components and seronegative antiphospholipid syndrome. Amniocentesis was performed due to fetal growth restriction (FGR). FISH with TUPLE1 (HIRA) probe was normal; however, chromosomal microarray identified a ~737 kb heterozygous loss between LCR22B-D. Subsequently, the same deletion was identified in the mother, which included CRKL and 19 other genes but excluded HIRA and TBX1, the typical candidate genes for 22q11.2DS pathogenesis. This case explores how loss of CRKL may contribute to immune dysregulation, as seen in the multiple severe autoimmune phenotypes of the mother, and FGR. Our experience confirms the importance of thorough workup in individuals with reduced penetrance of 22q11.2 DS features or atypical clinical presentations.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Síndrome de DiGeorge/genética , Retardo del Crecimiento Fetal/genética , Lupus Eritematoso Sistémico/genética , Adulto , Anticuerpos Antinucleares/sangre , Deleción Cromosómica , Cromosomas Humanos Par 22/genética , Síndrome de DiGeorge/sangre , Síndrome de DiGeorge/complicaciones , Síndrome de DiGeorge/patología , Femenino , Retardo del Crecimiento Fetal/sangre , Retardo del Crecimiento Fetal/diagnóstico , Retardo del Crecimiento Fetal/patología , Feto , Pruebas Genéticas , Haploinsuficiencia/genética , Humanos , Hibridación Fluorescente in Situ , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/complicaciones , Lupus Eritematoso Sistémico/patología , Madres , Penetrancia , Secuencias Repetitivas de Ácidos Nucleicos/genética
7.
Am J Med Genet A ; 185(5): 1519-1524, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33634591

RESUMEN

Three unrelated patients with similar microdeletions of chromosome 14q32.11 with shared phenotypes including language and developmental delay, and four overlapping genes -CALM1, TTC7B, PSMC1, and RPS6KA5 have been presented. All four genes are expressed in the brain and have haploinsufficiency scores, which reflect low tolerance to loss of function variation. An insight on the genes in the overlapping region, which may influence the resulting phenotype has been provided. Given the three patients' similar phenotypes and lack of normal variation in this region, it was suggested that this microdeletion may be associated with developmental and language delay.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/genética , Calmodulina/genética , Trastornos del Desarrollo del Lenguaje/genética , Proteínas/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Niño , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 14/genética , Hibridación Genómica Comparativa/métodos , Haploinsuficiencia/genética , Humanos , Trastornos del Desarrollo del Lenguaje/patología , Masculino , Linaje , Fenotipo
8.
Am J Hum Genet ; 99(5): 1015-1033, 2016 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-27745839

RESUMEN

In this exciting era of "next-gen cytogenetics," integrating genomic sequencing into the prenatal diagnostic setting is possible within an actionable time frame and can provide precise delineation of balanced chromosomal rearrangements at the nucleotide level. Given the increased risk of congenital abnormalities in newborns with de novo balanced chromosomal rearrangements, comprehensive interpretation of breakpoints could substantially improve prediction of phenotypic outcomes and support perinatal medical care. Herein, we present and evaluate sequencing results of balanced chromosomal rearrangements in ten prenatal subjects with respect to the location of regulatory chromatin domains (topologically associated domains [TADs]). The genomic material from all subjects was interpreted to be "normal" by microarray analyses, and their rearrangements would not have been detected by cell-free DNA (cfDNA) screening. The findings of our systematic approach correlate with phenotypes of both pregnancies with untoward outcomes (5/10) and with healthy newborns (3/10). Two pregnancies, one with a chromosomal aberration predicted to be of unknown clinical significance and another one predicted to be likely benign, were terminated prior to phenotype-genotype correlation (2/10). We demonstrate that the clinical interpretation of structural rearrangements should not be limited to interruption, deletion, or duplication of specific genes and should also incorporate regulatory domains of the human genome with critical ramifications for the control of gene expression. As detailed in this study, our molecular approach to both detecting and interpreting the breakpoints of structural rearrangements yields unparalleled information in comparison to other commonly used first-tier diagnostic methods, such as non-invasive cfDNA screening and microarray analysis, to provide improved genetic counseling for phenotypic outcome in the prenatal setting.


Asunto(s)
Aberraciones Cromosómicas , Anomalías Congénitas/genética , Reordenamiento Génico , Nucleótidos/genética , Diagnóstico Prenatal/métodos , Alelos , Mapeo Cromosómico , Anomalías Congénitas/diagnóstico , Femenino , Regulación de la Expresión Génica , Pruebas Genéticas , Genoma Humano , Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Cariotipificación , Masculino , Embarazo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Análisis de Secuencia de ADN , Translocación Genética
9.
Am J Hum Genet ; 96(3): 498-506, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25728775

RESUMEN

Chromatin remodeling through histone acetyltransferase (HAT) and histone deactylase (HDAC) enzymes affects fundamental cellular processes including the cell-cycle, cell differentiation, metabolism, and apoptosis. Nonsense mutations in genes that are involved in histone acetylation and deacetylation result in multiple congenital anomalies with most individuals displaying significant developmental delay, microcephaly and dysmorphism. Here, we report a syndrome caused by de novo heterozygous nonsense mutations in KAT6A (a.k.a., MOZ, MYST3) identified by clinical exome sequencing (CES) in four independent families. The same de novo nonsense mutation (c.3385C>T [p.Arg1129∗]) was observed in three individuals, and the fourth individual had a nearby de novo nonsense mutation (c.3070C>T [p.Arg1024∗]). Neither of these variants was present in 1,815 in-house exomes or in public databases. Common features among all four probands include primary microcephaly, global developmental delay including profound speech delay, and craniofacial dysmorphism, as well as more varied features such as feeding difficulties, cardiac defects, and ocular anomalies. We further demonstrate that KAT6A mutations result in dysregulation of H3K9 and H3K18 acetylation and altered P53 signaling. Through histone and non-histone acetylation, KAT6A affects multiple cellular processes and illustrates the complex role of acetylation in regulating development and disease.


Asunto(s)
Codón sin Sentido/genética , Discapacidades del Desarrollo/genética , Histona Acetiltransferasas/genética , Microcefalia/genética , Anomalías Múltiples/genética , Acetilación , Preescolar , Exoma , Femenino , Heterocigoto , Histona Acetiltransferasas/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Masculino , Mutación , Linaje
11.
Am J Med Genet A ; 176(12): 2829-2834, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30244526

RESUMEN

Nonimmune hydrops fetalis (NIHF) is a rare disorder with a high perinatal mortality of at least 50%. One cause of NIHF is generalized lymphatic dysplasia (GLD), a rare form of primary lymphedema of the extremities and systemic involvement including chylothoraces and pericardial effusions. An autosomal recessive form of GLD has been described, caused by variants in the PIEZO1 gene. It has been reported clinically to cause NIHF and childhood onset of facial and limb lymphedema, most of which were diagnosed postnatally. We present a case of a woman with recurrent pregnancies affected by NIHF because of novel compound heterozygous variants in the PIEZO1 gene diagnosed prenatally using exome sequencing (ES). Two variants in PIEZO1 (c.3206G>A and c.6208A>C) were identified that were inherited from the father and mother, and are predicted to cause a nonsense and missense change, respectively, in the PIEZO1 subunits. Ultrasound demonstrated severe bilateral pleural effusions, whole body edema and polyhydramnios. Histopathology revealed an increased number of lymphatic channels, many of which showed failure of luminal canalization. Sanger sequencing confirmed the same variants in a prior fetal demise. We provide phenotypic correlation with ultrasound and autopsy finding, review PIEZO1 variants as a cause of GLD and discuss the uses of prenatal ES to date.


Asunto(s)
Exoma , Variación Genética , Hidropesía Fetal/diagnóstico , Hidropesía Fetal/genética , Canales Iónicos/genética , Adulto , Autopsia , Biopsia , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Recién Nacido , Embarazo , Ultrasonografía Prenatal , Secuenciación del Exoma
12.
Hum Mol Genet ; 24(8): 2375-89, 2015 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-25574029

RESUMEN

Cardiac left ventricular outflow tract (LVOT) defects represent a common but heterogeneous subset of congenital heart disease for which gene identification has been difficult. We describe a 46,XY,t(1;5)(p36.11;q31.2)dn translocation carrier with pervasive developmental delay who also exhibited LVOT defects, including bicuspid aortic valve (BAV), coarctation of the aorta (CoA) and patent ductus arteriosus (PDA). The 1p breakpoint disrupts the 5' UTR of AHDC1, which encodes AT-hook DNA-binding motif containing-1 protein, and AHDC1-truncating mutations have recently been described in a syndrome that includes developmental delay, but not congenital heart disease [Xia, F., Bainbridge, M.N., Tan, T.Y., Wangler, M.F., Scheuerle, A.E., Zackai, E.H., Harr, M.H., Sutton, V.R., Nalam, R.L., Zhu, W. et al. (2014) De Novo truncating mutations in AHDC1 in individuals with syndromic expressive language delay, hypotonia, and sleep apnea. Am. J. Hum. Genet., 94, 784-789]. On the other hand, the 5q translocation breakpoint disrupts the 3' UTR of MATR3, which encodes the nuclear matrix protein Matrin 3, and mouse Matr3 is strongly expressed in neural crest, developing heart and great vessels, whereas Ahdc1 is not. To further establish MATR3 3' UTR disruption as the cause of the proband's LVOT defects, we prepared a mouse Matr3(Gt-ex13) gene trap allele that disrupted the 3' portion of the gene. Matr3(Gt-ex13) homozygotes are early embryo lethal, but Matr3(Gt-ex13) heterozygotes exhibit incompletely penetrant BAV, CoA and PDA phenotypes similar to those in the human proband, as well as ventricular septal defect (VSD) and double-outlet right ventricle (DORV). Both the human MATR3 translocation breakpoint and the mouse Matr3(Gt-ex13) gene trap insertion disturb the polyadenylation of MATR3 transcripts and alter Matrin 3 protein expression, quantitatively or qualitatively. Thus, subtle perturbations in Matrin 3 expression appear to cause similar LVOT defects in human and mouse.


Asunto(s)
Coartación Aórtica/genética , Válvula Aórtica/anomalías , Conducto Arterioso Permeable/genética , Enfermedades de las Válvulas Cardíacas/genética , Proteínas Asociadas a Matriz Nuclear/genética , Proteínas de Unión al ARN/genética , Adolescente , Animales , Coartación Aórtica/metabolismo , Válvula Aórtica/metabolismo , Enfermedad de la Válvula Aórtica Bicúspide , Preescolar , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Conducto Arterioso Permeable/metabolismo , Femenino , Silenciador del Gen , Enfermedades de las Válvulas Cardíacas/metabolismo , Ventrículos Cardíacos/anomalías , Ventrículos Cardíacos/metabolismo , Humanos , Recién Nacido , Masculino , Ratones , Mutagénesis Insercional , Proteínas Asociadas a Matriz Nuclear/metabolismo , Proteínas de Unión al ARN/metabolismo , Translocación Genética
13.
Hum Mutat ; 37(2): 148-54, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26507355

RESUMEN

Mandibulofacial dysostosis with microcephaly (MFDM) is a multiple malformation syndrome comprising microcephaly, craniofacial anomalies, hearing loss, dysmorphic features, and, in some cases, esophageal atresia. Haploinsufficiency of a spliceosomal GTPase, U5-116 kDa/EFTUD2, is responsible. Here, we review the molecular basis of MFDM in the 69 individuals described to date, and report mutations in 38 new individuals, bringing the total number of reported individuals to 107 individuals from 94 kindreds. Pathogenic EFTUD2 variants comprise 76 distinct mutations and seven microdeletions. Among point mutations, missense substitutions are infrequent (14 out of 76; 18%) relative to stop-gain (29 out of 76; 38%), and splicing (33 out of 76; 43%) mutations. Where known, mutation origin was de novo in 48 out of 64 individuals (75%), dominantly inherited in 12 out of 64 (19%), and due to proven germline mosaicism in four out of 64 (6%). Highly penetrant clinical features include, microcephaly, first and second arch craniofacial malformations, and hearing loss; esophageal atresia is present in an estimated ∼27%. Microcephaly is virtually universal in childhood, with some adults exhibiting late "catch-up" growth and normocephaly at maturity. Occasionally reported anomalies, include vestibular and ossicular malformations, reduced mouth opening, atrophy of cerebral white matter, structural brain malformations, and epibulbar dermoid. All reported EFTUD2 mutations can be found in the EFTUD2 mutation database (http://databases.lovd.nl/shared/genes/EFTUD2).


Asunto(s)
Anomalías Múltiples/genética , Pérdida Auditiva/genética , Discapacidad Intelectual/genética , Disostosis Mandibulofacial/genética , Microcefalia/genética , Mutación , Factores de Elongación de Péptidos/genética , Ribonucleoproteína Nuclear Pequeña U5/genética , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/patología , Secuencias de Aminoácidos , Bases de Datos Genéticas , Expresión Génica , Haploinsuficiencia , Pérdida Auditiva/diagnóstico , Pérdida Auditiva/patología , Humanos , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/patología , Disostosis Mandibulofacial/diagnóstico , Disostosis Mandibulofacial/patología , Microcefalia/diagnóstico , Microcefalia/patología , Modelos Moleculares , Datos de Secuencia Molecular , Penetrancia , Fenotipo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Empalme del ARN , Empalmosomas/genética
15.
BMC Med Genet ; 15: 49, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24886118

RESUMEN

BACKGROUND: Wiedemann-Steiner Syndrome (WSS) is characterized by short stature, a variety of dysmorphic facial and skeletal features, characteristic hypertrichosis cubiti (excessive hair on the elbows), mild-to-moderate developmental delay and intellectual disability. [MIM#: 605130]. Here we report two unrelated children for whom clinical exome sequencing of parent-proband trios was performed at UCLA, resulting in a molecular diagnosis of WSS and atypical clinical presentation. CASE PRESENTATION: For patient 1, clinical features at 9 years of age included developmental delay, craniofacial abnormalities, and multiple minor anomalies. Patient 2 presented at 1 year of age with developmental delay, microphthalmia, partial 3-4 left hand syndactyly, and craniofacial abnormalities. A de novo missense c.4342T>C variant and a de novo splice site c.4086+G>A variant were identified in the KMT2A gene in patients 1 and 2, respectively. CONCLUSIONS: Based on the clinical and molecular findings, both patients appear to have novel presentations of WSS. As the hallmark hypertrichosis cubiti was not initially appreciated in either case, this syndrome was not suspected during the clinical evaluation. This report expands the phenotypic spectrum of the clinical phenotypes and KMT2A variants associated with WSS.


Asunto(s)
Anomalías Múltiples/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Anomalías Múltiples/diagnóstico , Niño , Biología Computacional/métodos , Discapacidades del Desarrollo/diagnóstico , Exoma , Facies , Femenino , Variación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , N-Metiltransferasa de Histona-Lisina , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Masculino , Fenotipo , Síndrome
16.
Am J Med Genet A ; 164A(12): 3076-82, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25256560

RESUMEN

Deletions of chromosome 17q12 [OMIM 614527] encompass a wide range of phenotypes, including renal cysts, diabetes mellitus, pancreatic structural abnormalities, genital tract anomalies, developmental delay, learning difficulties, and more recently, autism spectrum disorder and schizophrenia. To date, gastrointestinal malformations have not been fully characterized in this syndrome. In this case report, we describe a four-year-old girl with a 17q12 microdeletion who was born with duodenal atresia, bilateral renal cysts, left kidney dysplasia, a midline cystic structure at the conus medullaris, and dysmorphic features. Both the patient and her affected father were found to have a deletion of 17q12, which encompasses the HNF1B (hepatocyte nuclear factor beta). It is hypothesized that HNF1B may play a role in intestinal differentiation and development. Our clinical report further expands the pre-and post-natal presentation of this rare microdeletion syndrome.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Múltiples/patología , Aberraciones Cromosómicas , Cromosomas Humanos Par 17/genética , Obstrucción Duodenal/genética , Factor Nuclear 1-beta del Hepatocito/deficiencia , Fenotipo , Femenino , Factor Nuclear 1-beta del Hepatocito/genética , Humanos , Atresia Intestinal , Análisis por Micromatrices , Síndrome
17.
JAMA ; 312(18): 1880-7, 2014 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-25326637

RESUMEN

IMPORTANCE: Clinical exome sequencing (CES) is rapidly becoming a common molecular diagnostic test for individuals with rare genetic disorders. OBJECTIVE: To report on initial clinical indications for CES referrals and molecular diagnostic rates for different indications and for different test types. DESIGN, SETTING, AND PARTICIPANTS: Clinical exome sequencing was performed on 814 consecutive patients with undiagnosed, suspected genetic conditions at the University of California, Los Angeles, Clinical Genomics Center between January 2012 and August 2014. Clinical exome sequencing was conducted as trio-CES (both parents and their affected child sequenced simultaneously) to effectively detect de novo and compound heterozygous variants or as proband-CES (only the affected individual sequenced) when parental samples were not available. MAIN OUTCOMES AND MEASURES: Clinical indications for CES requests, molecular diagnostic rates of CES overall and for phenotypic subgroups, and differences in molecular diagnostic rates between trio-CES and proband-CES. RESULTS: Of the 814 cases, the overall molecular diagnosis rate was 26% (213 of 814; 95% CI, 23%-29%). The molecular diagnosis rate for trio-CES was 31% (127 of 410 cases; 95% CI, 27%-36%) and 22% (74 of 338 cases; 95% CI, 18%-27%) for proband-CES. In cases of developmental delay in children (<5 years, n = 138), the molecular diagnosis rate was 41% (45 of 109; 95% CI, 32%-51%) for trio-CES cases and 9% (2 of 23, 95% CI, 1%-28%) for proband-CES cases. The significantly higher diagnostic yield (P value = .002; odds ratio, 7.4 [95% CI, 1.6-33.1]) of trio-CES was due to the identification of de novo and compound heterozygous variants. CONCLUSIONS AND RELEVANCE: In this sample of patients with undiagnosed, suspected genetic conditions, trio-CES was associated with higher molecular diagnostic yield than proband-CES or traditional molecular diagnostic methods. Additional studies designed to validate these findings and to explore the effect of this approach on clinical and economic outcomes are warranted.


Asunto(s)
Exoma , Enfermedades Genéticas Congénitas/diagnóstico , Técnicas de Diagnóstico Molecular , Enfermedades Raras/diagnóstico , Adolescente , Adulto , Niño , Preescolar , Discapacidades del Desarrollo/genética , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Mutación , Enfermedades Raras/genética , Análisis de Secuencia de ADN/métodos
18.
Am J Med Genet A ; 161A(5): 1167-72, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23532965

RESUMEN

Pierre Robin sequence (PRS) is a malformation pattern characterized by the core triad of retrognathia, glossoptosis, and cleft palate that causes difficulty in glossopharyngeal-laryngeal-vagal functions. The etiology of PRS remains largely unknown; previous reports have suggested that it is caused by intrauterine constriction or external conditions such as oligohydramnios, breech position, or abnormal uterine anatomy. Genetic causes include occurrence as a manifestation of many single gene conditions and chromosomal rearrangements. Positional effect on some loci or genes, including SOX9 has also been posited as a cause. Here, we report on an 18-month-old girl born with isolated PRS. Clinical chromosome microarray analysis (CMA) revealed a maternally inherited ~623 kb microdeletion that is -725 kb upstream of 5' SOX9 at chromosome locus 17q24.3. Her mother had cleft palate. This region, although devoid of any genes, is known to have a position effect on SOX9 due to elimination of highly conserved non-coding cis-regulatory elements. This report supports the evidence that deregulation of an intact SOX9 coding region is a cause of or associated with isolated PRS, and provides further evidence that CMA in the clinical setting is a powerful tool in detecting microdeletions in gene "desert" regions that have pathogenic position effect on specific genes.


Asunto(s)
Fibromatosis Gingival/genética , Hipertricosis/genética , Síndrome de Pierre Robin/genética , Factor de Transcripción SOX9/genética , Deleción Cromosómica , Cromosomas Humanos Par 17/genética , Femenino , Humanos , Lactante , Análisis de Secuencia por Matrices de Oligonucleótidos
19.
Am J Med Genet A ; 161A(8): 1985-91, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23794175

RESUMEN

Cat-Eye syndrome (CES), (OMIM 115470) also known as chromosome 22 partial tetrasomy or inverted duplicated 22q11, was first reported by Haab [1879] based on the primary features of eye coloboma and anal atresia. However, >60% of the patients lack these primary features. Here, we present a 9-month-old female who at birth was noted to have multiple defects, including facial asymmetry with asymmetric retrognathia, bilateral mandibular hypoplasia, branchial cleft sinus, right-sided muscular torticollis, esotropia, and an atretic right ear canal with low-to-moderate sensorineural hearing loss, bilateral preauricular ear tag/pits, and two skin tags on her left cheek. There were no signs of any colobomas or anal atresia. Hemifacial microsomia (HFM) was suspected clinically. Chromosome studies and FISH identified an extra marker originated from 22q11 consistent with CES, and this was confirmed by aCGH. This report expands the phenotypic variability of CES and includes partial tetrasomy of 22q11.1-q11.21 in the differential diagnosis of HFM. In addition, our case as well as the previous association of 22q11.2 deletions and duplications with facial asymmetry and features of HFM, supports the hypothesis that this chromosome region harbors genes important in the regulation of body plan symmetry, and in particular facial harmony.


Asunto(s)
Trastornos de los Cromosomas/complicaciones , Cromosomas Humanos Par 22/genética , Asimetría Facial/etiología , Adulto , Aneuploidia , Trastornos de los Cromosomas/genética , Hibridación Genómica Comparativa , Anomalías del Ojo , Femenino , Humanos , Hibridación Fluorescente in Situ , Lactante , Fenotipo
20.
Am J Med Genet A ; 161A(7): 1773-8, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23696469

RESUMEN

Deletions of the long arm of chromosome 18 have been previously reported in many patients. Most cases involve the more distal regions of the long arm (18q21.1->qter). However, proximal interstitial deletions involving 18q11.2 are extremely rare. Here we report on a 14-month-old female with a 4.7 Mb (19,667,062-24,401,876 hg19) de novo interstitial deletion within chromosomal band 18q11.2, which includes GATA6 and 24 other RefSeq genes. The clinical features of our patient include complex congenital heart defects, a double outlet right ventricle, a subaortic ventricular septal defect, D-malposed great arteries, an atrial septal defect, a dysplastic aortic valve and patent ductus arteriosus. In addition, she had renal anomalies-a duplicated collecting system on the left and mild right hydronephrosis. These heart and renal defects are not reported in other patients with 18q proximal interstitial deletions. Heterozygous point mutations in GATA6, encoding for a zinc finger transcription factor, have been shown to cause congenital heart defects. Given the well-established biological role of GATA6 in cardiac development, a deletion of GATA6 is very likely responsible for our patient's complex congenital heart defects. This is the smallest and most proximal 18q11.2 deletion involving GATA6 that is associated with complex congenital heart disease and renal anomalies.


Asunto(s)
Trastornos de los Cromosomas/genética , Cardiopatías Congénitas/genética , Riñón/anomalías , Deleción Cromosómica , Trastornos de los Cromosomas/etiología , Cromosomas Humanos Par 18/genética , Femenino , Factor de Transcripción GATA6/genética , Humanos , Lactante , Recién Nacido , Embarazo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA