Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Am J Hum Genet ; 98(6): 1146-1158, 2016 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-27259050

RESUMEN

Ataxia-pancytopenia (AP) syndrome is characterized by cerebellar ataxia, variable hematologic cytopenias, and predisposition to marrow failure and myeloid leukemia, sometimes associated with monosomy 7. Here, in the four-generation family UW-AP, linkage analysis revealed four regions that provided the maximal LOD scores possible, one of which was in a commonly microdeleted chromosome 7q region. Exome sequencing identified a missense mutation (c.2640C>A, p.His880Gln) in the sterile alpha motif domain containing 9-like gene (SAMD9L) that completely cosegregated with disease. By targeted sequencing of SAMD9L, we subsequently identified a different missense mutation (c.3587G>C, p.Cys1196Ser) in affected members of the first described family with AP syndrome, Li-AP. Neither variant is reported in the public databases, both affect highly conserved amino acid residues, and both are predicted to be damaging. With time in culture, lymphoblastic cell lines (LCLs) from two affected individuals in family UW-AP exhibited copy-neutral loss of heterozygosity for large portions of the long arm of chromosome 7, resulting in retention of only the wild-type SAMD9L allele. Newly established LCLs from both individuals demonstrated the same phenomenon. In addition, targeted capture and sequencing of SAMD9L in uncultured blood DNA from both individuals showed bias toward the wild-type allele. These observations indicate in vivo hematopoietic mosaicism. The hematopoietic cytopenias that characterize AP syndrome and the selective advantage for clones that have lost the mutant allele support the postulated role of SAMD9L in the regulation of cell proliferation. Furthermore, we show that AP syndrome is distinct from the dyskeratoses congenita telomeropathies, with which it shares some clinical characteristics.


Asunto(s)
Ataxia Cerebelosa/genética , Aberraciones Cromosómicas , Mutación Missense/genética , Pancitopenia/genética , Proteínas/genética , Adolescente , Adulto , Ataxia Cerebelosa/patología , Niño , Cromosomas Humanos Par 7/genética , Exoma/genética , Femenino , Ligamiento Genético , Genotipo , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Péptidos y Proteínas de Señalización Intracelular , Pérdida de Heterocigocidad , Masculino , Persona de Mediana Edad , Pancitopenia/patología , Linaje , Proteínas Supresoras de Tumor/genética , Adulto Joven
2.
Am J Med Genet A ; 164A(8): 2091-6, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24810580

RESUMEN

In 10 cases of 2p15p16.1 microdeletions reported worldwide to date, shared phenotypes included growth retardation, craniofacial and skeletal dysmorphic traits, internal organ defects, intellectual disability, nonverbal or low verbal status, abnormal muscle tone, and gross motor delays. The size of the deletions ranged from 0.3 to 5.7 Mb, where the smallest deletion involved the BCL11A, PAPOLG, and REL genes. Here we report on an 11-year-old male with a heterozygous de novo 0.2 Mb deletion containing a single gene, BCL11A, and a phenotype characterized by childhood apraxia of speech and dysarthria in the presence of general oral and gross motor dyspraxia and hypotonia as well as expressive language and mild intellectual delays. BCL11A is situated within the dyslexia susceptibility candidate region 3 (DYX3) candidate region on chromosome 2. The present case is the first to involve a single gene within the microdeletion region and a phenotype restricted to a subset of the traits observed in other cases with more extensive deletions.


Asunto(s)
Proteínas Portadoras/genética , Deleción Cromosómica , Estudios de Asociación Genética , Trastornos del Lenguaje/diagnóstico , Trastornos del Lenguaje/genética , Proteínas Nucleares/genética , Niño , Mapeo Cromosómico , Cromosomas Humanos Par 2 , Heterocigoto , Humanos , Masculino , Fenotipo , Proteínas Represoras , Índice de Severidad de la Enfermedad , Trastorno Fonológico
3.
Am J Med Genet B Neuropsychiatr Genet ; 165B(4): 345-56, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24807833

RESUMEN

Dyslexia, or specific reading disability, is a common developmental disorder that affects 5-12% of school-aged children. Dyslexia and its component phenotypes, assessed categorically or quantitatively, have complex genetic bases. The ability to rapidly name letters, numbers, and colors from rows presented visually correlates strongly with reading in multiple languages and is a valid predictor of reading and spelling impairment. Performance on measures of rapid naming and switching, RAN and RAS, is stable throughout elementary school years, with slowed performance persisting in adults who still manifest dyslexia. Targeted analyses of dyslexia candidate regions have included RAN measures, but only one other genome-wide linkage study has been reported. As part of a broad effort to identify genetic contributors to dyslexia, we performed combined oligogenic segregation and linkage analyses of measures of RAN and RAS in a family-based cohort ascertained through probands with dyslexia. We obtained strong evidence for linkage of RAN letters to the DYX3 locus on chromosome 2p and RAN colors to chromosome 10q, but were unable to confirm the chromosome 6p21 linkage detected for a composite measure of RAN colors and objects in the previous genome-wide study.


Asunto(s)
Cognición , Dislexia/genética , Estudio de Asociación del Genoma Completo , Lenguaje , Matemática , Sitios de Carácter Cuantitativo/genética , Lectura , Adolescente , Teorema de Bayes , Niño , Segregación Cromosómica/genética , Color , Intervalos de Confianza , Ligamiento Genético , Humanos
4.
Hum Mutat ; 34(12): 1672-8, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24027063

RESUMEN

PHARC (polyneuropathy, hearing loss, ataxia, retinitis pigmentosa, and cataracts) is a recently described autosomal-recessive neurodegenerative disease caused by mutations in the α-ß-hydrolase domain-containing 12 gene (ABHD12). Only five homozygous ABHD12 mutations have been reported and the pathogenesis of PHARC remains unclear. We evaluated a woman who manifested short stature as well as the typical features of PHARC. Sequence analysis of ABHD12 revealed a novel heterozygous c.1129A>T (p.Lys377*) mutation. Targeted comparative genomic hybridization detected a 59-kb deletion that encompasses exon 1 of ABHD12 and exons 1-4 of an adjacent gene, GINS1, and includes the promoters of both genes. The heterozygous deletion was also carried by the patient's asymptomatic mother. Quantitative reverse transcription-PCR demonstrated ∼50% decreased expression of ABHD12 RNA in lymphoblastoid cell lines from both individuals. Activity-based protein profiling of serine hydrolases revealed absence of ABHD12 hydrolase activity in the patient and 50% reduction in her mother. This is the first report of compound heterozygosity in PHARC and the first study to describe how a mutation might affect ABHD12 expression and function. The possible involvement of haploinsufficiency for GINS1, a DNA replication complex protein, in the short stature of the patient and her mother requires further studies.


Asunto(s)
Ataxia/genética , Catarata/genética , Monoacilglicerol Lipasas/genética , Mutación , Polineuropatías/genética , Retinitis Pigmentosa/genética , Adulto , Ataxia/diagnóstico , Ataxia/metabolismo , Catarata/diagnóstico , Catarata/metabolismo , Femenino , Expresión Génica , Orden Génico , Heterocigoto , Humanos , Masculino , Monoacilglicerol Lipasas/metabolismo , Linaje , Fenotipo , Polineuropatías/diagnóstico , Polineuropatías/metabolismo , Retinitis Pigmentosa/diagnóstico , Retinitis Pigmentosa/metabolismo , Eliminación de Secuencia , Transcripción Genética
5.
Am J Hum Genet ; 84(5): 692-7, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19409521

RESUMEN

We have established strong linkage evidence that supports mapping autosomal-dominant sensory/motor neuropathy with ataxia (SMNA) to chromosome 7q22-q32. SMNA is a rare neurological disorder whose phenotype encompasses both the central and the peripheral nervous system. In order to identify a gene responsible for SMNA, we have undertaken a comprehensive genomic evaluation of the region of linkage, including evaluation for repeat expansion and small deletions or duplications, capillary sequencing of candidate genes, and massively parallel sequencing of all coding exons. We excluded repeat expansion and small deletions or duplications as causative, and through microarray-based hybrid capture and massively parallel short-read sequencing, we identified a nonsynonymous variant in the human interferon-related developmental regulator gene 1 (IFRD1) as a disease-causing candidate. Sequence conservation, animal models, and protein structure evaluation support the involvement of IFRD1 in SMNA. Mutation analysis of IFRD1 in additional patients with similar phenotypes is needed for demonstration of causality and further evaluation of its importance in neurological diseases.


Asunto(s)
Ataxia/genética , Cromosomas Humanos Par 7/genética , Predisposición Genética a la Enfermedad , Neuropatía Hereditaria Motora y Sensorial/genética , Proteínas Inmediatas-Precoces/genética , Humanos , Mutación , Linaje
6.
Am J Med Genet B Neuropsychiatr Genet ; 159B(1): 61-71, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22095694

RESUMEN

Structural variations in the chromosome 22q11.2 region mediated by nonallelic homologous recombination result in 22q11.2 deletion (del22q11.2) and 22q11.2 duplication (dup22q11.2) syndromes. The majority of del22q11.2 cases have facial and cardiac malformations, immunologic impairments, specific cognitive profile and increased risk for schizophrenia and autism spectrum disorders (ASDs). The phenotype of dup22q11.2 is frequently without physical features but includes the spectrum of neurocognitive abnormalities. Although there is substantial evidence that haploinsufficiency for TBX1 plays a role in the physical features of del22q11.2, it is not known which gene(s) in the critical 1.5 Mb region are responsible for the observed spectrum of behavioral phenotypes. We identified an individual with a balanced translocation 46,XY,t(1;22)(p36.1;q11.2) and a behavioral phenotype characterized by cognitive impairment, autism, and schizophrenia in the absence of congenital malformations. Using somatic cell hybrids and comparative genomic hybridization (CGH) we mapped the chromosome-22 breakpoint within intron 7 of the GNB1L gene. Copy number evaluations and direct DNA sequencing of GNB1L in 271 schizophrenia and 513 autism cases revealed dup22q11.2 in two families with autism and private GNB1L missense variants in conserved residues in three families (P = 0.036). The identified missense variants affect residues in the WD40 repeat domains and are predicted to have deleterious effects on the protein. Prior studies provided evidence that GNB1L may have a role in schizophrenia. Our findings support involvement of GNB1L in ASDs as well.


Asunto(s)
Trastorno Autístico/genética , Predisposición Genética a la Enfermedad , Péptidos y Proteínas de Señalización Intracelular/genética , Adolescente , Secuencia de Bases , Estudios de Casos y Controles , Niño , Preescolar , Rotura Cromosómica , Variaciones en el Número de Copia de ADN/genética , Análisis Mutacional de ADN , Familia , Femenino , Duplicación de Gen/genética , Humanos , Recién Nacido , Cariotipificación , Masculino , Datos de Secuencia Molecular , Mutación/genética , Mutación Missense/genética , Linaje , Translocación Genética
7.
Behav Genet ; 41(1): 31-42, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20852926

RESUMEN

Dyslexia is a complex learning disability with evidence for a genetic basis. Strategies that may be useful for dissecting its genetic basis include the study of component phenotypes, which may simplify the underlying genetic complexity, and use of an analytic approach that accounts for the multilocus nature of the trait to guide the investigation and increase power to detect individual loci. Here we present results of a genetic analysis of spelling disability as a component phenotype. Spelling disability is informative in analysis of extended pedigrees because it persists into adulthood. We show that a small number of hypothesized loci are sufficient to explain the inheritance of the trait in our sample, and that each of these loci maps to one of four genomic regions. Individual trait models and locations are a function of whether a verbal IQ adjustment is included, suggesting mediation through both IQ-related and unrelated pathways.


Asunto(s)
Mapeo Cromosómico , Dislexia/genética , Estudios de Asociación Genética , Inteligencia/genética , Modelos Genéticos , Fenotipo , Aprendizaje Verbal , Logro , Adolescente , Niño , Dislexia/psicología , Femenino , Genotipo , Humanos , Masculino , Herencia Multifactorial , Sitios de Carácter Cuantitativo
8.
J Leukoc Biol ; 110(5): 829-837, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34061398

RESUMEN

Triggering receptor expressed on myeloid cells 2 (TREM2) is an immunoglobulin-like receptor expressed by certain myeloid cells, such as macrophages, dendritic cells, osteoclasts, and microglia. In the brain, TREM2 plays an important role in the immune function of microglia, and its dysfunction is linked to various neurodegenerative conditions in humans. Ablation of TREM2 or its adaptor protein TYROBP causes polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (also known as Nasu-Hakola disorder) with early onset of dementia, whereas some missense variants in TREM2 are associated with an increased risk of late-onset Alzheimer's disease. The human TREM2 gene is subject to alternative splicing, and its major, full-length canonic transcript encompasses 5 exons. Herein, we report a novel alternatively spliced TREM2 isoform without exon 2 (Δe2), which constitutes a sizable fraction of TREM2 transcripts and has highly variable inter-individual expression in the human brain (average frequency 10%; range 3.7-35%). The protein encoded by Δe2 lacks a V-set immunoglobulin domain from its extracellular part but retains its transmembrane and cytoplasmic domains. We demonstrated Δe2 protein expression in TREM2-positive THP-1 cells, in which the expression of full-length transcript was precluded by CRISPR/Cas9 disruption of the exon 2 coding frame. Similar to the full-length TREM2, Δe2 is sorted to the plasma membrane and is subject to receptor shedding. In "add-back" experiments, Δe2 TREM2 had diminished capacity to restore phagocytosis of amyloid beta peptide and promote IFN-I response as compared to full-length TREM2. Our findings suggest that changes in the balance of two mutually exclusive TREM2 isoforms may modify the dosage of full-length transcript potentially weakening some TREM2 receptor functions in the human brain.


Asunto(s)
Empalme Alternativo/fisiología , Encéfalo/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Humanos , Dominios de Inmunoglobulinas , Fagocitosis/genética , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo
9.
Mov Disord ; 25(10): 1409-17, 2010 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-20629132

RESUMEN

The parkinsonian syndromes comprise a highly heterogeneous group of disorders. Although 15 loci are linked to predominantly familial Parkinson's disease (PD), additional PD loci are likely to exist. We recently identified a multigenerational family of Danish and German descent in which five males in three generations presented with a unique syndrome characterized by parkinsonian features and variably penetrant spasticity for which X-linked disease transmission was strongly suggested (XPDS). Autopsy in one individual failed to reveal synucleinopathy; however, there was a significant four-repeat tauopathy in the striatum. Our objective was to identify the locus responsible for this unique parkinsonian disorder. Members of the XPDS family were genotyped for markers spanning the X chromosome. Two-point and multipoint linkage analyses were performed and the candidate region refined by analyzing additional markers. A multipoint LOD(max) score of 2.068 was obtained between markers DXS991 and DXS993. Haplotype examination revealed an approximately 20 cM region bounded by markers DXS8042 and DXS1216 that segregated with disease in all affected males and obligate carrier females and was not carried by unaffected at-risk males. To reduce the possibility of a false-positive linkage result, multiple loci and genes associated with other parkinsonian or spasticity syndromes were excluded. In conclusion, we have identified a unique X-linked parkinsonian syndrome with variable spasticity and four-repeat tau pathology, and defined a novel candidate gene locus spanning approximately 28 Mb from Xp11.2-Xq13.3.


Asunto(s)
Cromosomas Humanos X/genética , Genes Ligados a X/genética , Predisposición Genética a la Enfermedad , Repeticiones de Microsatélite/genética , Enfermedad de Parkinson/complicaciones , Tauopatías/genética , Adulto , Anciano , Anciano de 80 o más Años , Análisis Mutacional de ADN , Salud de la Familia , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Tauopatías/complicaciones
10.
Front Immunol ; 11: 559342, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33101276

RESUMEN

The R47H variant in the microglial triggering receptor expressed on myeloid cell 2 (TREM2) receptor is a strong risk factor for Alzheimer's disease (AD). To characterize processes affected by R47H, we performed an integrative network analysis of genes expressed in brains of AD patients with R47H, sporadic AD without the variant, and patients with polycystic lipomembranous osteodysplasia with sclerosing leukoencephalopathy (PLOSL), systemic disease with early-onset dementia caused by loss-of-function mutations in TREM2 or its adaptor TYRO protein tyrosine kinase-binding protein (TYROBP). Although sporadic AD had few perturbed microglial and immune genes, TREM2 R47H AD demonstrated upregulation of interferon type I response and pro-inflammatory cytokines accompanied by induction of NKG2D stress ligands. In contrast, PLOSL had distinct sets of highly perturbed immune and microglial genes that included inflammatory mediators, immune signaling, cell adhesion, and phagocytosis. TREM2 knockout (KO) in THP1, a human myeloid cell line that constitutively expresses the TREM2- TYROBP receptor, inhibited response to the viral RNA mimetic poly(I:C) and phagocytosis of amyloid-beta oligomers; overexpression of ectopic TREM2 restored these functions. Compared with wild-type protein, R47H TREM2 had a higher stimulatory effect on the interferon type I response signature. Our findings point to a role of the TREM2 receptor in the control of the interferon type I response in myeloid cells and provide insight regarding the contribution of R47H TREM2 to AD pathology.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Inmunidad , Glicoproteínas de Membrana/genética , Mutación , Receptores Inmunológicos/genética , Alelos , Enfermedad de Alzheimer/patología , Sustitución de Aminoácidos , Biomarcadores , Biopsia , Encéfalo/patología , Línea Celular , Biología Computacional/métodos , Citocinas/metabolismo , Expresión Génica , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Mutación con Pérdida de Función , Glicoproteínas de Membrana/metabolismo , Fagocitosis/genética , Fagocitosis/inmunología , Receptores Inmunológicos/metabolismo , Transducción de Señal
11.
Am J Med Genet B Neuropsychiatr Genet ; 150B(4): 570-4, 2009 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-18980218

RESUMEN

We previously reported a five-generation family manifesting an autosomal dominant disorder of facial myokymia and dystonic/choreic movements (FDFM). The dyskinetic episodes are initially paroxysmal but may become constant. With increasing age they may lessen or even disappear. The previous study excluded nine candidate genes chosen for their association with myokymia or chorea and two regions containing single or clustered ion channel genes. We now report identification by whole genome linkage analysis of a broad region on chromosome 3p21-3q21 that segregates with the disease in all 10 affected members in three generations who participated in the study. GENEHUNTER-MODSCORE Version 2.0.1 provided a maximum multipoint LOD score of 3.099. No other disorders primarily characterized by myokymia, dystonia, or chorea are known to map to this region. Identification of additional families with FDFM may narrow the critical region and facilitate the choice of candidate genes for further analysis.


Asunto(s)
Cromosomas Humanos Par 3/genética , Discinesias/genética , Enfermedades del Nervio Facial/genética , Mapeo Cromosómico , Estudios de Seguimiento , Ligamiento Genético , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad , Humanos , Linaje
12.
Behav Genet ; 38(5): 462-75, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18607713

RESUMEN

To understand the genetic architecture of dyslexia and identify the locations of genes involved, we performed linkage analyses in multigenerational families using a phonological memory phenotype--Nonword Repetition (NWR). A genome scan was first performed on 438 people from 51 families (DS-1) and linkage was assessed using variance components (VC), Bayesian oligogenic (BO), and parametric analyses. For replication, the genome scan and analyses were repeated on 693 people from 93 families (DS-2). For the combined set (DS-C), analyses were performed with all three methods in the regions that were identified in both samples. In DS-1, regions on chromosomes 4p, 6q, 12p, 17q, and 22q exceeded our initial threshold for linkage, with 17q providing a parametric LOD score of 3.2. Analysis with DS-2 confirmed the locations on chromosomes 4p and 12p. The strongest VC and BO signals in both samples were on chromosome 4p in DS-C, with a parametric multipoint LOD(max) of 2.36 for the 4p locus. Our linkage analyses of NWR in dyslexia provide suggestive and reproducible evidence for linkage to 4p12 and 12p in both samples, and significant evidence for linkage to 17q in one of the samples. These results warrant further studies of phonological memory and chromosomal regions identified here in other datasets.


Asunto(s)
Dislexia/genética , Genoma , Memoria , Adulto , Niño , Salud de la Familia , Femenino , Ligamiento Genético , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Masculino , Persona de Mediana Edad , Padres , Fenotipo
13.
Am J Med Genet A ; 146A(14): 1880-5, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18553554

RESUMEN

The Waardenburg syndromes (WS) account for approximately 2% of congenital sensorineural deafness. This heterogeneous group of diseases currently can be categorized into four major subtypes (WS types 1-4) on the basis of characteristic clinical features. Multiple genes have been implicated in WS, and mutations in some genes can cause more than one WS subtype. In addition to eye, hair, and skin pigmentary abnormalities, dystopia canthorum and broad nasal bridge are seen in WS type 1. Mutations in the PAX3 gene are responsible for the condition in the majority of these patients. In addition, mutations in PAX3 have been found in WS type 3 that is distinguished by musculoskeletal abnormalities, and in a family with a rare subtype of WS, craniofacial-deafness-hand syndrome (CDHS), characterized by dysmorphic facial features, hand abnormalities, and absent or hypoplastic nasal and wrist bones. Here we describe a woman who shares some, but not all features of WS type 3 and CDHS, and who also has abnormal cranial bones. All sinuses were hypoplastic, and the cochlea were small. No sequence alteration in PAX3 was found. These observations broaden the clinical range of WS and suggest there may be genetic heterogeneity even within the CDHS subtype.


Asunto(s)
Anomalías Craneofaciales/genética , Pérdida Auditiva Sensorineural/genética , Síndrome de Waardenburg/diagnóstico , Síndrome de Waardenburg/genética , Adulto , Anomalías Craneofaciales/patología , ADN/genética , Análisis Mutacional de ADN , Femenino , Pérdida Auditiva Sensorineural/congénito , Humanos , Factor de Transcripción PAX3 , Factores de Transcripción Paired Box/genética , Senos Paranasales/anomalías , Fenotipo , Síndrome de Waardenburg/clasificación
14.
Biomark Genes ; 1(1)2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30854516

RESUMEN

Students without specific learning disabilities [SLDs] [n=18] and with one of three persisting SLDs in written language despite early and current specialized instruction-Dysgraphia [n=21], Dyslexia [n=40], or oral and written language learning disability OWL LD [n=14]- in grades 4 to 9 [N=56 boys, 38 girls] completed behavioral phenotyping assessment and gave a small blood or saliva sample. Molecular analyses informed by current cross-site research on gene candidates for learning disabilities identified associations between molecular genetic markers and the two defining behavioral phenotypes for each SLDs-WL; dysgraphia [impaired writing alphabet from memory for rs3743204 and sentence copying in best handwriting for rs79382 both in DYX1C1], dyslexia [impaired silent word reading/decoding rate for rs4535189 in DCDC2 and impaired spelling/encoding for rs374205 in DYX1C1], and OWL LD [impaired aural syntax comprehension for rs807701 and oral syntax construction for rs807701 both in DYX1C1]. Implications of these identified associations between molecular markers for alleles for different sites within two gene candidates [and mostly one] and hallmark phenotypes are discussed for translation science [application to practice] and neuroimaging that has identified contrasting brain bases for each of the three SLDs.

15.
PLoS One ; 11(4): e0153864, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27120335

RESUMEN

Childhood apraxia of speech (CAS) is a severe and socially debilitating form of speech sound disorder with suspected genetic involvement, but the genetic etiology is not yet well understood. Very few known or putative causal genes have been identified to date, e.g., FOXP2 and BCL11A. Building a knowledge base of the genetic etiology of CAS will make it possible to identify infants at genetic risk and motivate the development of effective very early intervention programs. We investigated the genetic etiology of CAS in two large multigenerational families with familial CAS. Complementary genomic methods included Markov chain Monte Carlo linkage analysis, copy-number analysis, identity-by-descent sharing, and exome sequencing with variant filtering. No overlaps in regions with positive evidence of linkage between the two families were found. In one family, linkage analysis detected two chromosomal regions of interest, 5p15.1-p14.1, and 17p13.1-q11.1, inherited separately from the two founders. Single-point linkage analysis of selected variants identified CDH18 as a primary gene of interest and additionally, MYO10, NIPBL, GLP2R, NCOR1, FLCN, SMCR8, NEK8, and ANKRD12, possibly with additive effects. Linkage analysis in the second family detected five regions with LOD scores approaching the highest values possible in the family. A gene of interest was C4orf21 (ZGRF1) on 4q25-q28.2. Evidence for previously described causal copy-number variations and validated or suspected genes was not found. Results are consistent with a heterogeneous CAS etiology, as is expected in many neurogenic disorders. Future studies will investigate genome variants in these and other families with CAS.


Asunto(s)
Apraxias/genética , Variaciones en el Número de Copia de ADN/genética , Predisposición Genética a la Enfermedad/genética , Habla/fisiología , Exoma/genética , Femenino , Ligamiento Genético/genética , Genotipo , Humanos , Escala de Lod , Masculino , Linaje , Riesgo
16.
Arch Neurol ; 62(4): 597-600, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15824259

RESUMEN

BACKGROUND: Paroxysmal nonkinesigenic dyskinesia (PNKD) is a rare disorder characterized by attacks of involuntary movements brought on by stress, alcohol, or caffeine, but not by movement. An autosomal dominant form of this disorder was mapped to chromosome 2q33-36, and different missense mutations in exon 1 of the myofibrillogenesis regulator 1 (MR1) gene were identified recently in 2 kindreds. OBJECTIVES: To describe studies on a new pedigree with PNKD, to explore the possibility of locus heterogeneity, and to further delineate the spectrum of mutations in MR1 in 2 families with PNKD. DESIGN, SETTING, AND PATIENTS: All 10 exons of MR1 were sequenced in DNA from members of 2 pedigrees with autosomal dominant PNKD. RESULTS: Different missense mutations in exon 1 of MR1 that cosegregate with disease were identified in each multiplex family. These single-nucleotide mutations predicted substitution of valine for alanine in residue 7 in one family and residue 9 in the other. The same mutations were found in the only 2 families previously published. Family history and haplotype analysis make it unlikely that the families with the same mutations are related. CONCLUSIONS: The function of MR1 is unknown, but the 2 mutations identified in the 4 families with PNKD studied to date are predicted to disrupt the amino terminal alpha-helix suggesting that this region of the gene is critical for proper gene function under stressful conditions. Study of additional families will be important to determine whether analysis of a single exon (MR1 exon 1) is sufficient for genetic testing purposes.


Asunto(s)
Sustitución de Aminoácidos/genética , Corea/genética , Cromosomas Humanos Par 2/genética , Predisposición Genética a la Enfermedad/genética , Proteínas Musculares/genética , Mutación Missense/genética , Alanina/genética , Alanina/metabolismo , Corea/metabolismo , Corea/fisiopatología , Mapeo Cromosómico , Análisis Mutacional de ADN , Exones/genética , Femenino , Genes Dominantes , Marcadores Genéticos/genética , Pruebas Genéticas , Haplotipos , Humanos , Masculino , Proteínas Musculares/química , Proteínas Musculares/metabolismo , Linaje , Estructura Secundaria de Proteína/genética , Valina/genética , Valina/metabolismo
17.
JAMA Neurol ; 72(8): 920-7, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26076170

RESUMEN

IMPORTANCE: The R47H variant in the triggering receptor expressed on myeloid cells 2 gene (TREM2), a modulator of the immune response of microglia, is a strong genetic risk factor for Alzheimer disease (AD) and possibly other neurodegenerative disorders. OBJECTIVE: To investigate a large family with late-onset AD (LOAD), in which R47H cosegregated with 75% of cases. DESIGN, SETTING, AND PARTICIPANTS: This study includes genetic and pathologic studies of families with LOAD from 1985 to 2014. A total of 131 families with LOAD (751 individuals) were included from the University of Washington Alzheimer Disease Research Center. To identify LOAD genes/risk factors in the LOAD123 family with 21 affected members and 12 autopsies, we sequenced 4 exomes. Candidate variants were tested for cosegregation with the disease. TREM2 R47H was genotyped in an additional 130 families with LOAD. We performed clinical and neuropathological assessments of patients with and without R47H and evaluated the variant's effect on brain pathology, cellular morphology, and expression of microglial markers. MAIN OUTCOMES AND MEASURES: We assessed the effect of TREM2 genotype on age at onset and disease duration. We compared Braak and Consortium to Establish a Registry for Alzheimer's Disease scores, presence of α-synuclein and TAR DNA-binding protein 43 aggregates, and additional vascular or Parkinson pathology in TREM2 R47H carriers vs noncarriers. Microglial activation was assessed by quantitative immunohistochemistry and morphometry. RESULTS: Twelve of 16 patients with AD in the LOAD123 family carried R47H. Eleven patients with dementia had apolipoprotein E 4 (ApoE4) and R47H genotypes. We also found a rare missense variant, D353N, in a nominated AD risk gene, unc-5 homolog C (UNC5C), in 5 affected individuals in the LOAD123 family. R47H carriers demonstrated a shortened disease duration (mean [SD], 6.7 [2.8] vs 11.1 [6.6] years; 2-tailed t test; P = .04) and more frequent α-synucleinopathy. The panmicroglial marker ionized calcium-binding adapter molecule 1 was decreased in all AD cases and the decrease was most pronounced in R47H carriers (mean [SD], in the hilus: 0.114 [0.13] for R47H_AD vs 0.574 [0.26] for control individuals; 2-tailed t test; P = .005 and vs 0.465 [0.32] for AD; P = .02; in frontal cortex gray matter: 0.006 [0.004] for R47H_AD vs 0.016 [0.01] for AD; P = .04 and vs 0.033 [0.013] for control individuals; P < .001). Major histocompatibility complex class II, a marker of microglial activation, was increased in all patients with AD (AD: 2.5, R47H_AD: 2.7, and control: 1.0; P < .01). CONCLUSIONS AND RELEVANCE: Our results demonstrate a complex genetic landscape of LOAD, even in a single pedigree with an apparent autosomal dominant pattern of inheritance. ApoE4, TREM2 R47H, and rare variants in other genes, such as UNC5C D353N, are likely responsible for the notable occurrence of AD in this family. Our findings support the role of the TREM2 receptor in microglial clearance of aggregation-prone proteins that is compromised in R47H carriers and may accelerate the course of disease.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Encéfalo/patología , Glicoproteínas de Membrana/genética , Receptores Inmunológicos/genética , Edad de Inicio , Anciano , Anciano de 80 o más Años , Exoma , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Genotipo , Humanos , Masculino
18.
Arch Neurol ; 59(8): 1291-5, 2002 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12164726

RESUMEN

BACKGROUND: The autosomal dominant spinocerebellar ataxias (SCAs) are a clinically and genetically heterogeneous group of neurodegenerative disorders. Although molecular genetic studies have so far implicated 16 loci in the etiology of these diseases, approximately 30% of families with SCAs remain unlinked. OBJECTIVES: To report the location of a gene causing a "pure" autosomal dominant cerebellar ataxia in one family and to describe the clinical phenotype. PATIENTS: We have identified a 4-generation American family of English and Dutch ethnicity with a pure cerebellar ataxia displaying an autosomal dominant pattern of inheritance. The disease typically has its onset in the third and fourth decades of life, shows no evidence of anticipation, progresses slowly, and does not appear to decrease life expectancy. Clinical DNA testing excluded SCA1, 2, 3, 6, 7, and 8. METHODS: A genome-wide linkage analysis at a 10 centimorgan (cM) level was performed with samples from 26 family members (11 affected, 10 clinically unaffected at risk, and 5 spouses). RESULTS: Assuming 90% penetrance, we found suggestive evidence of linkage to chromosome 19, with a lod score of 2.49 for D19S571. More detailed mapping in this region provided a maximum 2-point lod score of 2.57 at theta = 0 for D19S254 and a maximum multipoint lod score of 4.72 at D19S926. By haplotype construction a 22-cM critical region from D19S601 to the q telomere was defined. CONCLUSIONS: We have mapped a gene for an autosomal dominant SCA to chromosome 19q13.4-qter in one family. The critical region overlaps with the locus for SCA14, a disease described in a single Japanese family and characterized by axial myoclonus. Myoclonus was not seen in the family we studied, but it remains possible that the 2 disorders are allelic variants.


Asunto(s)
Cromosomas Humanos Par 19 , Ligamiento Genético , Adolescente , Adulto , Niño , Mapeo Cromosómico , Femenino , Genes Dominantes , Humanos , Masculino , Persona de Mediana Edad , Linaje , Ataxias Espinocerebelosas
19.
Am J Med Genet ; 114(4): 450-7, 2002 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-11992570

RESUMEN

The autosomal dominant (AD) spinocerebellar ataxias (SCAs) and hereditary sensory neuropathies (HSN) are heterogeneous disorders characterized by variable clinical, electrophysiological, and neuropathological profiles. The SCAs are clinically characterized by slowly progressive incoordination of gait often associated with poor coordination of hands, speech, and eyes. Peripheral neuropathy is not a frequent part of the SCA syndrome. In contrast, the HSNs are primarily characterized by progressive sensory loss. There is substantial clinical overlap between the various SCAs and the various HSNs, and they often cannot be differentiated on the basis of clinical or neuro-imaging studies. We have identified a five-generation American family of Irish ancestry with a unique neurological disorder displaying an AD pattern of inheritance. There was variable expressivity and severity of symptoms including sensory loss, ataxia, pyramidal tract signs, and muscle weakness. Nerve conduction studies were consistent with a sensory axonal neuropathy. Muscle biopsy revealed neurogenic atrophy and brain MRI showed mild cerebellar atrophy. To identify the responsible locus we pursued a whole genome linkage analysis. After analyzing 114 markers, linkage to D7S486 was detected with a two point LOD score of 4.79 at theta = 0.00. Evaluation of additional markers in the region provided a maximum LOD score of 6.36 at theta = 0.00 for marker D7S2554. Haplotype analysis delimited an approximately 14-cM region at 7q22-q32 between markers D7S2418 and D7S1804 cosegregating with the disease. Because this disorder does not easily fall into either the SCA or HSN categories, it is designated sensory/motor neuropathy with ataxia (SMNA).


Asunto(s)
Ataxia/genética , Cromosomas Humanos Par 7 , Adolescente , Adulto , Anciano , Ataxia/clasificación , Ataxia/patología , Encéfalo/patología , Ataxia Cerebelosa/genética , Niño , Mapeo Cromosómico , Genes Dominantes , Humanos , Escala de Lod , Imagen por Resonancia Magnética , Persona de Mediana Edad , Datos de Secuencia Molecular , Atrofia Muscular/genética , Linaje
20.
Psychiatr Genet ; 22(5): 226-34, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22517379

RESUMEN

OBJECTIVES: The aim of this pilot study was to investigate a measure of motor sequencing deficit as a potential endophenotype of speech sound disorder (SSD) in a multigenerational family with evidence of familial SSD. METHODS: In a multigenerational family with evidence of a familial motor-based SSD, affectation status and a measure of motor sequencing during oral motor testing were obtained. To further investigate the role of motor sequencing as an endophenotype for genetic studies, parametric and nonparametric linkage analyses were carried out using a genome-wide panel of 404 microsatellites. RESULTS: In seven of the 10 family members with available data, SSD affectation status and motor sequencing status coincided. Linkage analysis revealed four regions of interest, 6p21, 7q32, 7q36, and 8q24, primarily identified with the measure of motor sequencing ability. The 6p21 region overlaps with a locus implicated in rapid alternating naming in a recent genome-wide dyslexia linkage study. The 7q32 locus contains a locus implicated in dyslexia. The 7q36 locus borders on a gene known to affect the component traits of language impairment. CONCLUSION: The results are consistent with a motor-based endophenotype of SSD that would be informative for genetic studies. The linkage results in this first genome-wide study in a multigenerational family with SSD warrant follow-up in additional families and with fine mapping or next-generation approaches to gene identification.


Asunto(s)
Endofenotipos , Composición Familiar , Ligamiento Genético , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Trastornos del Lenguaje/genética , Niño , Preescolar , Femenino , Humanos , Escala de Lod , Masculino , Linaje , Trastorno Fonológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA