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

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Psychiatry ; 28(4): 1647-1663, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36117209

RESUMEN

Childhood apraxia of speech (CAS), the prototypic severe childhood speech disorder, is characterized by motor programming and planning deficits. Genetic factors make substantive contributions to CAS aetiology, with a monogenic pathogenic variant identified in a third of cases, implicating around 20 single genes to date. Here we aimed to identify molecular causation in 70 unrelated probands ascertained with CAS. We performed trio genome sequencing. Our bioinformatic analysis examined single nucleotide, indel, copy number, structural and short tandem repeat variants. We prioritised appropriate variants arising de novo or inherited that were expected to be damaging based on in silico predictions. We identified high confidence variants in 18/70 (26%) probands, almost doubling the current number of candidate genes for CAS. Three of the 18 variants affected SETBP1, SETD1A and DDX3X, thus confirming their roles in CAS, while the remaining 15 occurred in genes not previously associated with this disorder. Fifteen variants arose de novo and three were inherited. We provide further novel insights into the biology of child speech disorder, highlighting the roles of chromatin organization and gene regulation in CAS, and confirm that genes involved in CAS are co-expressed during brain development. Our findings confirm a diagnostic yield comparable to, or even higher, than other neurodevelopmental disorders with substantial de novo variant burden. Data also support the increasingly recognised overlaps between genes conferring risk for a range of neurodevelopmental disorders. Understanding the aetiological basis of CAS is critical to end the diagnostic odyssey and ensure affected individuals are poised for precision medicine trials.


Asunto(s)
Apraxias , Trastornos del Habla , Niño , Humanos , Trastornos del Habla/genética , Apraxias/genética , Mapeo Cromosómico , Causalidad , Encéfalo , N-Metiltransferasa de Histona-Lisina
2.
Blood ; 138(4): 304-317, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-33786586

RESUMEN

Hematopoiesis is extrinsically controlled by cells of the bone marrow microenvironment, including skeletal lineage cells. The identification and subsequent studies of distinct subpopulations of maturing skeletal cells is currently limited because of a lack of methods to isolate these cells. We found that murine Lin-CD31-Sca-1-CD51+ cells can be divided into 4 subpopulations by using flow cytometry based on their expression of the platelet-derived growth factor receptors ⍺ and ß (PDGFR⍺ and PDGFRß). The use of different skeletal lineage reporters confirmed the skeletal origin of the 4 populations. Multiplex immunohistochemistry studies revealed that all 4 populations were localized near the growth plate and trabecular bone and were rarely found near cortical bone regions or in central bone marrow. Functional studies revealed differences in their abundance, colony-forming unit-fibroblast capacity, and potential to differentiate into mineralized osteoblasts or adipocytes in vitro. Furthermore, the 4 populations had distinct gene expression profiles and differential cell surface expression of leptin receptor (LEPR) and vascular cell adhesion molecule 1 (VCAM-1). Interestingly, we discovered that 1 of these 4 different skeletal populations showed the highest expression of genes involved in the extrinsic regulation of B lymphopoiesis. This cell population varied in abundance between distinct hematopoietically active skeletal sites, and significant differences in the proportions of B-lymphocyte precursors were also observed in these distinct skeletal sites. This cell population also supported pre-B lymphopoiesis in culture. Our method of isolating 4 distinct maturing skeletal populations will help elucidate the roles of distinct skeletal niche cells in regulating hematopoiesis and bone.


Asunto(s)
Linfocitos B/inmunología , Diferenciación Celular/inmunología , Linfopoyesis/inmunología , Músculo Esquelético/inmunología , Animales , Diferenciación Celular/genética , Linfopoyesis/genética , Ratones , Ratones Transgénicos
3.
Am J Med Genet A ; 191(2): 357-369, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36349505

RESUMEN

Fragile X syndrome (FXS) is caused by hypermethylation of the FMR1 promoter due to the full mutation expansion (full mutation [FM]: CGG ≥ 200 repeats) and silencing of FMR1. Assessment of mosaicism for active-unmethylated alleles has prognostic utility. This study examined relationships between FMR1 methylation in different tissues with FMR1 messenger ribonucleic acid (mRNA) and intellectual functioning in 87 males with FXS (1.89-43.17 years of age). Methylation sensitive Southern blot (mSB) and Methylation Specific-Quantitative Melt Aanalysis (MS-QMA) were used to examine FMR1 methylation. FMR1 mRNA levels in blood showed strong relationships with FMR1 methylation assessed using MS-QMA in blood (n = 68; R2  = 0.597; p = 1.4 × 10-10 ) and buccal epithelial cells (BEC) (n = 62; R2  = 0.24; p = 0.003), with these measures also showing relationships with intellectual functioning scores (p < 0.01). However, these relationships were not as strong for mSB, with ~40% of males with only FM alleles that were 100% methylated and non-mosaic by mSB, showing methylation mosaicism by MS-QMA. This was confirmed through presence of detectable levels of FMR1 mRNA in blood. In summary, FMR1 methylation levels in blood and BEC examined by MS-QMA were significantly associated with FMR1 mRNA levels and intellectual functioning in males with FXS. These relationships were not as strong for mSB, which underestimated prevalence of mosaicism.


Asunto(s)
Síndrome del Cromosoma X Frágil , Masculino , Humanos , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/genética , Mosaicismo , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Metilación de ADN/genética , Mutación , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Am J Med Genet A ; 188(1): 304-309, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34545686

RESUMEN

The FMR1 premutation (PM:55-199 CGG) is associated with fragile X-associated tremor/ataxia syndrome (FXTAS) and when maternally transmitted is at risk of expansion to a hypermethylated full mutation (FM: ≥ 200 CGG) that causes fragile X syndrome (FXS). We describe a maternally transmitted PM (77 CGG) that was passed to a son (103 CGG), and to a daughter (220-1822 CGG), who were affected with FXTAS and FXS, respectively. The male with the PM showed low-level mosaicism for normal size of 30 and 37 CGG. This male had two offspring: one female mosaic for PM and FM (56, 157, >200 CGG) and another with only a 37 CGG allele detected in multiple tissues, neither with a clinical phenotype. The female with the 37 CGG allele showed normal levels of FMR1 methylation and mRNA and passed this 37 CGG allele to one of her daughters, who was also unaffected. These findings show that post-zygotic paternal retraction can lead to low-level mosaicism for normal size alleles, with these normal alleles being functional when passed over two generations.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Síndrome del Cromosoma X Frágil , Alelos , Metilación de ADN , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/genética , Humanos , Masculino , Mutación , Expansión de Repetición de Trinucleótido
5.
Am J Med Genet A ; 185(5): 1498-1503, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33544979

RESUMEN

Fragile X syndrome (FXS) is caused by CGG expansions of ≥200 repeats (full mutation: FM). Typically, FM causes abnormal methylation of the FMR1 promoter and silencing of FMR1, leading to reduction of FMRP, a protein essential for normal neurodevelopment. However, if unmethylated, these alleles cause over-expression of FMR1 mRNA which has been associated with Fragile X Tremor and Ataxia Syndrome (FXTAS), a late onset disorder. This report details the molecular and clinical profile of an asymptomatic male (29 years) identified as a result of cascade testing who was found to have a rare unmethylated FM (UFM) allele, as well as premutation (PM: 55-199 CGG) size alleles in multiple tissues. Full-scale IQ was within the normal range and minimal features of autism were observed. Southern blot analysis identified FM smears in blood (220-380 CGG) and saliva (212-378 CGG). A PM of 159 CGG was identified in blood and saliva. FMR1 promoter methylation analysis showed all alleles to be unmethylated. FMR1 mRNA levels were greater than fivefold of median levels in typically developing controls and males with FXS mosaic for PM and FM alleles. Issues raised during genetic counseling related to risk for FXTAS associated with UFM and elevated FMR1 mRNA levels, as well as, reproductive options, with implications for future practice.


Asunto(s)
Ataxia/genética , Trastorno Autístico/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Temblor/genética , Adulto , Alelos , Ataxia/patología , Trastorno Autístico/fisiopatología , Metilación de ADN/genética , Síndrome del Cromosoma X Frágil/patología , Tamización de Portadores Genéticos , Humanos , Masculino , Mutación/genética , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Temblor/patología , Expansión de Repetición de Trinucleótido/genética , Adulto Joven
6.
Int J Mol Sci ; 21(20)2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-33086711

RESUMEN

Fragile X syndrome (FXS) is a leading single-gene cause of intellectual disability (ID) with autism features. This study analysed diagnostic and prognostic utility of the Fragile X-Related Epigenetic Element 2 DNA methylation (FREE2m) assessed by Methylation Specific-Quantitative Melt Analysis and the EpiTYPER system, in retrospectively retrieved newborn blood spots (NBS) and newly created dried blood spots (DBS) from 65 children with FXS (~2-17 years). A further 168 NBS from infants from the general population were used to establish control reference ranges, in both sexes. FREE2m analysis showed sensitivity and specificity approaching 100%. In FXS males, NBS FREE2m strongly correlated with intellectual functioning and autism features, however associations were not as strong for FXS females. Fragile X mental retardation 1 gene (FMR1) mRNA levels in blood were correlated with FREE2m in both NBS and DBS, for both sexes. In females, DNAm was significantly increased at birth with a decrease in childhood. The findings support the use of FREE2m analysis in newborns for screening, diagnostic and prognostic testing in FXS.


Asunto(s)
Trastorno Autístico/genética , Metilación de ADN/genética , Síndrome del Cromosoma X Frágil/genética , Discapacidad Intelectual/genética , Adolescente , Niño , Preescolar , Estudios de Cohortes , Epigénesis Genética , Femenino , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Humanos , Lactante , Recién Nacido , Masculino , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo
8.
Int J Mol Sci ; 20(16)2019 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-31405222

RESUMEN

Although fragile X syndrome (FXS) is caused by a hypermethylated full mutation (FM) expansion with ≥200 cytosine-guanine-guanine (CGG) repeats, and a decrease in FMR1 mRNA and its protein (FMRP), incomplete silencing has been associated with more severe autism features in FXS males. This study reports on brothers (B1 and B2), aged 5 and 2 years, with autistic features and language delay, but a higher non-verbal IQ in comparison to typical FXS. CGG sizing using AmplideX PCR only identified premutation (PM: 55-199 CGGs) alleles in blood. Similarly, follow-up in B1 only revealed PM alleles in saliva and skin fibroblasts; whereas, an FM expansion was detected in both saliva and buccal DNA of B2. While Southern blot analysis of blood detected an unmethylated FM, methylation analysis with a more sensitive methodology showed that B1 had partially methylated PM alleles in blood and fibroblasts, which were completely unmethylated in buccal and saliva cells. In contrast, B2 was partially methylated in all tested tissues. Moreover, both brothers had FMR1 mRNA ~5 fold higher values than those of controls, FXS and PM cohorts. In conclusion, the presence of unmethylated FM and/or PM in both brothers may lead to an overexpression of toxic expanded mRNA in some cells, which may contribute to neurodevelopmental problems, including elevated autism features.


Asunto(s)
Trastorno Autístico/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , ARN Mensajero/genética , Alelos , Preescolar , Metilación de ADN , Humanos , Masculino , Mosaicismo , Mutación , Hermanos , Regulación hacia Arriba
9.
Nat Genet ; 37(3): 254-64, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15696166

RESUMEN

Transcriptional repression of methylated genes can be mediated by the methyl-CpG binding protein MeCP2. Here we show that human Brahma (Brm), a catalytic component of the SWI/SNF-related chromatin-remodeling complex, associates with MeCP2 in vivo and is functionally linked with repression. We used a number of different molecular approaches and chromatin immunoprecipitation strategies to show a unique cooperation between Brm, BAF57 and MeCP2. We show that Brm and MeCP2 assembly on chromatin occurs on methylated genes in cancer and the gene FMR1 in fragile X syndrome. These experimental findings identify a new role for SWI/SNF in gene repression by MeCP2.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Cromosómicas no Histona/fisiología , Proteínas de Unión al ADN/fisiología , Silenciador del Gen/fisiología , Proteínas Represoras/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Animales , Proteínas de Drosophila , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , Histonas/fisiología , Humanos , Proteína 2 de Unión a Metil-CpG , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Proteínas de Unión al ARN/genética
10.
J Biol Chem ; 287(5): 3156-64, 2012 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-22167198

RESUMEN

The highly conserved DYNLL1 (LC8) protein was originally discovered as a light chain of the dynein motor complex, but is increasingly emerging as a sequence-specific regulator of protein dimerization with hundreds of targets and wide-ranging cellular functions. Despite its important roles, DYNLL1's own regulation remains poorly understood. Here we identify ASCIZ (ATMIN/ZNF822), an essential Zn(2+) finger protein with dual roles in the DNA base damage response and as a developmental transcription factor, as a conserved regulator of Dynll1 gene expression. DYNLL1 levels are reduced by ∼10-fold in the absence of ASCIZ in human, mouse and chicken cells. ASCIZ binds directly to the Dynll1 promoter and regulates its activity in a Zn(2+) finger-dependent manner. DYNLL1 protein in turn interacts with ten binding sites in the ASCIZ transcription activation domain, and high DYNLL1 levels inhibit the transcriptional activity of ASCIZ. In addition, DYNLL1 was also required for DNA damage-induced ASCIZ focus formation. The dual ability of ASCIZ to activate Dynll1 gene expression and to sense free DYNLL1 protein levels enables a simple dynamic feedback loop to adjust DYNLL1 levels to cellular needs. The ASCIZ-DYNLL1 feedback loop represents a novel mechanism for auto-regulation of gene expression, where the gene product directly inhibits the transcriptional activator while bound at its own promoter.


Asunto(s)
Proteínas Portadoras/metabolismo , Dineínas Citoplasmáticas/biosíntesis , Regulación Enzimológica de la Expresión Génica/fisiología , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas/fisiología , Zinc/metabolismo , Animales , Sitios de Unión , Proteínas Portadoras/genética , Línea Celular , Pollos , Dineínas Citoplasmáticas/genética , Humanos , Ratones , Proteínas Nucleares/genética , Factores de Transcripción , Transcripción Genética/fisiología , Dedos de Zinc
11.
Am J Pathol ; 180(2): 811-8, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22155108

RESUMEN

Bone marrow (BM) fibrosis is a feature of severe hyperparathyroidism. Consistent with this observation, mice expressing constitutively active parathyroid hormone (PTH)/PTH-related peptide receptors (PPR) in osteoblasts (PPR*Tg) display BM fibrosis. To obtain insight into the nature of BM fibrosis in such a model, a double-mutant mouse expressing constitutively active PPR and green fluorescent protein (GFP) under the control of the type I collagen promoter (PPR*Tg/GFP) was generated. Confocal microscopy and flow cytometry revealed the presence of a cell population expressing GFP (GFP(+)) that was also positive for the hematopoietic marker CD45 in the BM of both PPR*Tg/GFP and control animals. This cell population was expanded in PPR*Tg/GFP. The existence of cells expressing both type I collagen and CD45 in the adult BM was confirmed by IHC and fluorescence-activated cell sorting. An analysis of total RNA extracted from sorted GFP(+)CD45(+) cells showed that these cells produced type I collagen and PTH/PTH-related peptide receptor and receptor activator for NF-κB mRNAs, further supporting their features of being both mesenchymal and hematopoietic lineages. Similar cells, known as fibrocytes, are also present in pathological fibroses. Our findings, thus, indicate that the BM is a permissive microenvironment for the differentiation of fibrocyte-like cells and raise the possibility that these cells could contribute to the pathogenesis of BM fibrosis.


Asunto(s)
Biomarcadores/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Mielofibrosis Primaria/patología , Animales , Médula Ósea/metabolismo , Diferenciación Celular , Colágeno Tipo I , Proteínas Fluorescentes Verdes/metabolismo , Ratones , Ratones Transgénicos , Osteoblastos/metabolismo , Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea/metabolismo , Mielofibrosis Primaria/metabolismo , Receptor de Hormona Paratiroídea Tipo 1/metabolismo
12.
Arterioscler Thromb Vasc Biol ; 32(8): 1910-6, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22723437

RESUMEN

OBJECTIVE: The postural tachycardia syndrome (POTS) has multiple symptoms, chief among which are tachycardia, weakness, and recurrent blackouts while standing. Previous research has implicated dysfunction of the norepinephrine transporter. A coding mutation in the norepinephrine transporter gene (SLC6A2) sequence has been reported in 1 family kindred only. The goal of the present study was to further characterize the role and regulation of the SLC6A2 gene in POTS. METHODS AND RESULTS: Sympathetic nervous system responses to head-up tilt were examined by combining norepinephrine plasma kinetics measurements and muscle sympathetic nerve activity recordings in patients with POTS compared with that in controls. The SLC6A2 gene sequence was investigated in leukocytes from POTS patients and healthy controls using single nucleotide polymorphisms genotyping, bisulphite sequencing, and chromatin immunoprecipitation assays for histone modifications and binding of the transcriptional regulatory complex, methyl-CpG binding protein 2. The expression of norepinephrine transporter was lower in POTS patients compared with healthy volunteers. In the absence of altered SLC6A2 gene sequence or promoter methylation, this reduced expression was directly correlated with chromatin modifications. CONCLUSIONS: We propose that chromatin-modifying events associated with SLC6A2 gene suppression may constitute a mechanism of POTS.


Asunto(s)
Epigénesis Genética , Proteínas de Transporte de Noradrenalina a través de la Membrana Plasmática/genética , Síndrome de Taquicardia Postural Ortostática/genética , Adulto , Metilación de ADN , Femenino , Perfilación de la Expresión Génica , Hemodinámica , Humanos , Masculino , Polimorfismo Genético , Síndrome de Taquicardia Postural Ortostática/fisiopatología , Regiones Promotoras Genéticas
13.
Disabil Health J ; 16(2): 101423, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36639256

RESUMEN

BACKGROUND: Angelman syndrome (AS) is a rare genetic condition characterized by global developmental delay, including severe intellectual disability. The parents of persons with AS experience increased stress, anxiety, and depression. This impacts parents' career choices and productivity. OBJECTIVE: To estimate, for the first time, the total productivity lost by the parents of persons with AS over a 10-year period in Australia and the corresponding cost to society. METHODS: A cost-of-illness model with simulated follow-up over a 10-year period was developed, with 2019 as the baseline year, facilitated by a Markov chain of life tables. The prevalence of persons with AS and their parents, the productivity-adjusted life years (PALYs) lost by parents, and the cost to society were estimated. Key data were obtained from a prospective cohort of AS families, peer-reviewed literature, and publicly available sources. RESULTS: The base-case productivity burden borne by the estimated 330 living parents of the 428 prevalent persons with AS totaled AUD$45.30 million, corresponding to a loss of 38.42% of PALYs per parent. CONCLUSIONS: Caring for a child with AS has a significant impact on the productivity of affected parents, with a large associated impact on the broader Australian economy.


Asunto(s)
Síndrome de Angelman , Personas con Discapacidad , Niño , Humanos , Australia/epidemiología , Años de Vida Ajustados por Calidad de Vida , Estudios Prospectivos , Padres , Costo de Enfermedad
14.
Turk J Gastroenterol ; 34(7): 736-746, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37232463

RESUMEN

BACKGROUND/AIMS: Cancer studies suffer from an overestimation of prediction of survival when both recurrence and death are of interest. This longitudinal study aimed to mitigate this problem utilizing a semi-competing risk approach evaluating the factors affecting recurrence and postoperative death in patients with colorectal cancer. MATERIALS AND METHODS: This longitudinal prospective study was conducted in 284 patients with resected colorectal cancer who were referred to the Imam Khomeini Clinic in Hamadan, Iran, during 2001-2017. Primary outcomes were postoperative outcomes and patient survival, including time to recurrence (of colorectal cancer), time to death, and time to death after recurrence. All patients who were alive at the end of the study were censored for death and who did not experience recurrence of colorectal cancer were also censored for recurrent colorectal cancer. The relationship between underlying demographics and clinical factors and the outcomes was assessed using a semicompeting risk approach. RESULTS: The results of the multivariable analysis showed that having metastasis to other sites (hazard ratio = 36.03; 95% CI = 19.48- 66.64) and higher pathological node (pN) stage (hazard ratio = 2.46; 95% CI = 1.32-4.56) were associated with a raised hazard of recurrence. The fewer chemotherapies (hazard ratio = 0.39; 95% CI = 0.17-0.88) and higher pN stages (hazard ratio = 4.32; 95% CI = 1.27-14.75) showed significantly higher hazards of death without recurrence. Having metastasis to other sites (hazard ratio = 2.67; 95% CI = 1.24-5.74) and higher pN stages (hazard ratio = 1.91; 95% CI = 1.02-3.61) were linked with the higher hazard of death after recurrence. CONCLUSION: Considering findings on death /recu rrenc e-spe cific predictors obtained in this study to manage the outcomes in patients with colorectal cancer, tailored strategies for preventive and interventional plans should be deliberated.


Asunto(s)
Neoplasias Colorrectales , Recurrencia Local de Neoplasia , Humanos , Estadificación de Neoplasias , Estudios Longitudinales , Estudios Prospectivos , Factores de Riesgo , Neoplasias Colorrectales/cirugía , Neoplasias Colorrectales/patología , Estudios Retrospectivos
15.
Autism ; 27(7): 1891-1905, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-36691305

RESUMEN

LAY ABSTRACT: Autistic children experience increased the rates of sleep problems. These sleep problems have been associated with mother's mental health symptoms. However, the direction of these relationships is not well understood. This study investigated the relationships between autistic children's sleep problems and mothers' mental health over a 12-year period using data collected as part of the Longitudinal Study of Australian Children. Data from 397 autistic children and their mothers were included in this study. Mothers completed a questionnaire about their own mental health and common childhood sleep problems at four time points from 4-5 years to 14-15 years. The results showed important relationships between mothers' mental health symptoms and child sleep problems at two time points. Specifically, (1) mothers' mental health symptoms when the child was aged 4 to 5 years predicted child sleep problems at age 6 to 7 years; and (2) child sleep problems at age 12-13 years predicted mothers' mental health symptoms when the child was aged 14 to 15 years. Interestingly, these significant relationships also coincide with key developmental transition time points, when the child is transitioning in and out of primary school. These findings highlight the need for increased support for both the child and mother at these times to optimise outcomes for both.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Trastornos del Sueño-Vigilia , Femenino , Niño , Humanos , Adolescente , Salud Mental , Estudios Longitudinales , Trastorno Autístico/complicaciones , Trastorno Autístico/epidemiología , Australia/epidemiología , Madres/psicología , Relaciones Madre-Hijo , Trastornos del Sueño-Vigilia/epidemiología
16.
J Autism Dev Disord ; 53(4): 1682-1692, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34292487

RESUMEN

The study characterised differences in costs associated with raising a child between four rare disorders and examined the associations between these costs with clinical severity. Caregivers of 108 individuals with Prader-Willi, Angelman (AS), Chromosome 15q Duplication and fragile X (FXS) syndromes completed a modified Client Services Receipt Inventory and participants completed intellectual/developmental functioning and autism assessments. AS incurred the highest yearly costs per individual ($AUD96,994), while FXS had the lowest costs ($AUD33,221). Intellectual functioning negatively predicted total costs, after controlling for diagnosis. The effect of intellectual functioning on total costs for those with AS was significantly different to the other syndromes. The study highlights the significant costs associated with these syndromes, particularly AS, linked with severity of intellectual functioning.


Asunto(s)
Síndrome de Angelman , Trastorno del Espectro Autista , Síndrome del Cromosoma X Frágil , Síndrome de Prader-Willi , Niño , Humanos , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/genética , Síndrome de Prader-Willi/complicaciones , Cromosomas Humanos Par 15/genética , Trastorno del Espectro Autista/complicaciones , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/complicaciones , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Australia , Duplicación Cromosómica
17.
Eur J Med Genet ; 65(4): 104456, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35218942

RESUMEN

Angelman syndrome (AS) is a severe neurodevelopmental disorder caused by loss of expression of the maternally-inherited UBE3A on chromosome 15q11.2. In AS due to a chromosomal deletion that encompasses UBE3A, paternal uniparental disomy of chromosome 15, or imprinting defects (ImpD), the SNRPN locus is unmethylated, while in neurotypical individuals, it is ∼50% methylated. We present the developmental profile of two adults with mild AS assessed using standardized behavioral and neurodevelopmental measures. Both had intellectual disability with unusually advanced verbal communication skills compared to other individuals with AS. Methylation of the SNRPN locus was examined using Methylation Specific Quantitative Melt Analysis (MS-QMA) in different tissues at one time point for participant A (22 years) and two time points for participant B (T1: 22 years, T2: 25 years), and these levels were compared to a typical AS cohort. While participant A showed methylation levels comparable to the typical AS cohort, participant B showed methylation mosaicism in all tissues at both time points and changes in methylation levels from T1 to T2. AS should be considered in individuals with intellectual disability and verbal speech who may not have the typical symptoms of AS.


Asunto(s)
Síndrome de Angelman , Adulto , Síndrome de Angelman/genética , Cromosomas Humanos Par 15/genética , Metilación de ADN , Impresión Genómica , Humanos , Mosaicismo , Disomía Uniparental , Proteínas Nucleares snRNP/genética
18.
Res Dev Disabil ; 131: 104338, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36179574

RESUMEN

BACKGROUND: Despite the increasing number of clinical trials involving children with neurodevelopmental disorders, appropriate and objective outcome measures for behavioral symptoms are still required. AIM: This study assessed the agreement between parents' and clinical researchers' ratings of behavioral problem severity in children with fragile X syndrome (FXS) and chromosome 15 imprinting disorders. METHODS AND PROCEDURES: The cohort comprised 123 children (64% males), aged 3-17 years, with FXS (n = 79), Prader-Willi (PWS; n = 19), Angelman (AS; n = 15), and Chromosome 15q duplication (n = 10) syndromes. Specific items from the Autism Diagnostic Observation Schedule-Second Edition and Aberrant Behavior Checklist-Community Edition mapping to corresponding behavioral domains were selected ad-hoc, to assess behavioral problems. OUTCOMES AND RESULTS: Inter-rater agreement for the cohort was slight for self-injury (Intraclass Correlation Coefficient (ICC) = 0.12), fair for tantrums/aggression (0.24) and mannerisms/stereotypies (0.25), and moderate for hyperactivity (0.48). When stratified by diagnosis, ICC ranged from poor (0; self-injury, AS and PWS) to substantial (0.48; hyperactivity, females with FXS). CONCLUSIONS AND IMPLICATIONS: The high level of inter-rater disagreement across most domains suggests that parents' and researchers' assessments led to discrepant appraisal of behavioral problem severity. These findings have implications for treatment targets and outcome measure selection in clinical trials, supporting a multi-informant approach.


Asunto(s)
Síndrome del Cromosoma X Frágil , Síndrome de Prader-Willi , Problema de Conducta , Niño , Masculino , Femenino , Humanos , Síndrome del Cromosoma X Frágil/diagnóstico , Síndrome del Cromosoma X Frágil/genética , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/genética , Cromosomas Humanos Par 15/genética , Padres
19.
JAMA Netw Open ; 5(1): e2141911, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34982160

RESUMEN

Importance: Newborn screening for Angelman syndrome (AS), Prader-Willi syndrome (PWS), and chromosome 15 duplication syndrome (Dup15q) may lead to benefit from early diagnosis and treatment. Objective: To examine the feasibility of newborn screening for these chromosome 15 imprinting disorders at population scale. Design, Setting, and Participants: In this diagnostic study, the validation data set for the first-tier SNRPN test, called methylation-specific quantitative melt analysis (MS-QMA), included 109 PWS, 48 AS, 9 Dup15q, and 1190 population control newborn blood spots (NBS) and peripheral tissue samples from participants recruited from January 2000 to December 2016. The test data set included NBS samples from 16 579 infants born in 2011. Infants with an NBS identified as positive for PWS, AS, or Dup15q by the first-tier test were referred for droplet digital polymerase chain reaction, real-time polymerase chain reaction, and low-coverage whole-genome sequencing for confirmatory testing. Data analyses were conducted between February 12, 2015, and August 15, 2020. Results: In the validation data set, the median age for the 77 patients with PWS was 3.00 years (IQR, 0.01-44.50 years); for the 46 patients with AS, 2.76 years (IQR, 0.028 to 49.00 years); and for the 9 patients with Dup15q, 4.00 years (IQR, 1.00 to 28.00 years). Thirty-eight patients (51.4%) in the PWS group, 20 patients (45.5%) in the AS group, and 6 patients (66.7%) in the Dup15q group who had sex reported were male. The validation data set showed MS-QMA sensitivity of 99.0% for PWS, 93.8% for AS, and 77.8% for Dup15q; specificity of 100% for PWS, AS, and Dup15q; positive predictive and negative predictive values of 100% for PWS and AS; and a positive predictive value of 87.5% and negative predictive value of 100% for Dup15q. In the test data set of NBS samples from 16 579 infants, 92 had a positive test result using a methylation ratio cut-off of 3 standard deviations from the mean. Of these patients, 2 were confirmed to have PWS; 2, AS; and 1, maternal Dup15q. With the use of more conservative PWS- and AS-specific thresholds for positive calls from the validation data set, 9 positive NBS results were identified by MS-QMA in this cohort. The 2 PWS and 2 AS calls were confirmed by second-tier testing, but the 1 Dup15q case was not confirmed. Together, these results provided prevalence estimates of 1 in 8290 for both AS and PWS and 1 in 16 579 for maternal Dup15q, with positive predictive values for first-tier testing at 67.0% for AS, 33.0% for PWS, and 44.0% for combined detection of chromosome 15 imprinting disorders for the validation data set. Conclusions and Relevance: The findings of this diagnostic study suggest that it is feasible to screen for all chromosome 15 imprinting disorders using SNRPN methylation analysis, with 5 individuals identified with these disorders out of 16 579 infants screened.


Asunto(s)
Síndrome de Angelman , Cromosomas Humanos Par 15/genética , Pruebas Genéticas/métodos , Tamizaje Neonatal/métodos , Síndrome de Prader-Willi , Adolescente , Adulto , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Niño , Preescolar , Duplicación Cromosómica/genética , Metilación de ADN/genética , Estudios de Factibilidad , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Síndrome de Prader-Willi/diagnóstico , Síndrome de Prader-Willi/genética , Adulto Joven
20.
Autism ; 25(2): 490-501, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33092410

RESUMEN

LAY ABSTRACT: We investigated whether a commonly used research assessment - the Autism Observation Scale for Infants (AOSI) - accurately measures autism behaviours among infants showing early signs of autism identified within the community. The AOSI is often included in studies tracking the development of infants at increased likelihood of autism, such as the infant siblings of diagnosed children. However, the suitability of this measure has not previously been tested with community-referred infants. We administered the AOSI with infants when aged 9 to 14 months and again 6 months later. Our researchers - independent of the AOSI development team and newly trained on this measure - were able to administer the brief interactive assessment and score it accurately. The infants' AOSI scores were linked to their scores on other established and validated clinical assessments, particularly at the second visit when average age was 18 months. Stronger correspondence of AOSI and other scores at this second visit suggests early autism behaviours are better established and more consistent by 18 months of age, even though these infants showed clear enough signs of possible autism to prompt referral to our study around 12 months of age. However, the moderate association of AOSI scores over time suggests that, like infant siblings - who mostly do not develop autism - community-identified infants showing early signs may also have variable developmental pathways in early life.


Asunto(s)
Trastorno del Espectro Autista , Trastorno Autístico , Trastorno del Espectro Autista/diagnóstico , Trastorno Autístico/diagnóstico , Niño , Desarrollo Infantil , Humanos , Lactante , Hermanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA