Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Science ; 366(6464): 454-460, 2019 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-31624095

RESUMEN

We present histological and molecular analyses of the developing human cerebellum from 30 days after conception to 9 months after birth. Differences in developmental patterns between humans and mice include spatiotemporal expansion of both ventricular and rhombic lip primary progenitor zones to include subventricular zones containing basal progenitors. The human rhombic lip persists longer through cerebellar development than in the mouse and undergoes morphological changes to form a progenitor pool in the posterior lobule, which is not seen in other organisms, not even in the nonhuman primate the macaque. Disruptions in human rhombic lip development are associated with posterior cerebellar vermis hypoplasia and Dandy-Walker malformation. The presence of these species-specific neural progenitor populations refines our insight into human cerebellar developmental disorders.


Asunto(s)
Cerebelo/embriología , Cerebelo/crecimiento & desarrollo , Células Madre/citología , Animales , Síndrome de Dandy-Walker , Humanos , Ratones , Malformaciones del Sistema Nervioso , Análisis Espacio-Temporal , Especificidad de la Especie , Transcriptoma
2.
Am J Hum Genet ; 105(3): 606-615, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31474318

RESUMEN

Cerebellar malformations are diverse congenital anomalies frequently associated with developmental disability. Although genetic and prenatal non-genetic causes have been described, no systematic analysis has been performed. Here, we present a large-exome sequencing study of Dandy-Walker malformation (DWM) and cerebellar hypoplasia (CBLH). We performed exome sequencing in 282 individuals from 100 families with DWM or CBLH, and we established a molecular diagnosis in 36 of 100 families, with a significantly higher yield for CBLH (51%) than for DWM (16%). The 41 variants impact 27 neurodevelopmental-disorder-associated genes, thus demonstrating that CBLH and DWM are often features of monogenic neurodevelopmental disorders. Though only seven monogenic causes (19%) were identified in more than one individual, neuroimaging review of 131 additional individuals confirmed cerebellar abnormalities in 23 of 27 genetic disorders (85%). Prenatal risk factors were frequently found among individuals without a genetic diagnosis (30 of 64 individuals [47%]). Single-cell RNA sequencing of prenatal human cerebellar tissue revealed gene enrichment in neuronal and vascular cell types; this suggests that defective vasculogenesis may disrupt cerebellar development. Further, de novo gain-of-function variants in PDGFRB, a tyrosine kinase receptor essential for vascular progenitor signaling, were associated with CBLH, and this discovery links genetic and non-genetic etiologies. Our results suggest that genetic defects impact specific cerebellar cell types and implicate abnormal vascular development as a mechanism for cerebellar malformations. We also confirmed a major contribution for non-genetic prenatal factors in individuals with cerebellar abnormalities, substantially influencing diagnostic evaluation and counseling regarding recurrence risk and prognosis.


Asunto(s)
Cerebelo/anomalías , Cerebelo/diagnóstico por imagen , Estudios de Cohortes , Femenino , Humanos , Masculino , Embarazo
3.
Am J Hum Genet ; 105(3): 640-657, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31402090

RESUMEN

The identification of genetic variants implicated in human developmental disorders has been revolutionized by second-generation sequencing combined with international pooling of cases. Here, we describe seven individuals who have diverse yet overlapping developmental anomalies, and who all have de novo missense FBXW11 variants identified by whole exome or whole genome sequencing and not reported in the gnomAD database. Their phenotypes include striking neurodevelopmental, digital, jaw, and eye anomalies, and in one individual, features resembling Noonan syndrome, a condition caused by dysregulated RAS signaling. FBXW11 encodes an F-box protein, part of the Skp1-cullin-F-box (SCF) ubiquitin ligase complex, involved in ubiquitination and proteasomal degradation and thus fundamental to many protein regulatory processes. FBXW11 targets include ß-catenin and GLI transcription factors, key mediators of Wnt and Hh signaling, respectively, critical to digital, neurological, and eye development. Structural analyses indicate affected residues cluster at the surface of the loops of the substrate-binding domain of FBXW11, and the variants are predicted to destabilize the protein and/or its interactions. In situ hybridization studies on human and zebrafish embryonic tissues demonstrate FBXW11 is expressed in the developing eye, brain, mandibular processes, and limb buds or pectoral fins. Knockdown of the zebrafish FBXW11 orthologs fbxw11a and fbxw11b resulted in embryos with smaller, misshapen, and underdeveloped eyes and abnormal jaw and pectoral fin development. Our findings support the role of FBXW11 in multiple developmental processes, including those involving the brain, eye, digits, and jaw.


Asunto(s)
Encéfalo/anomalías , Anomalías del Ojo/genética , Dedos/anomalías , Mutación Missense , Fenotipo , Ubiquitina-Proteína Ligasas/genética , Proteínas con Repetición de beta-Transducina/genética , Adolescente , Adulto , Niño , Preescolar , Femenino , Humanos , Masculino
4.
Birth Defects Res ; 110(5): 443-455, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29316344

RESUMEN

BACKGROUND: MC1R, a G-protein coupled receptor with high affinity for alpha-melanocyte stimulating hormone (αMSH), modulates pigment production in melanocytes from many species and is associated with human melanoma risk. MC1R mutations affecting human skin and hair color also have pleiotropic effects on the immune response and analgesia. Variants affecting human pigmentation in utero alter the congenital phenotype of both oculocutaneous albinism and congenital melanocytic naevi, and have a possible effect on birthweight. METHODS AND RESULTS: By in situ hybridization, RT-PCR and immunohistochemistry, we show that MC1R is widely expressed during human, chick and mouse embryonic and fetal stages in many somatic tissues, particularly in the musculoskeletal and nervous systems, and conserved across evolution in these three amniotes. Its dynamic pattern differs from that of TUBB3, a gene overlapping the same locus in humans and encoding class III ß-tubulin. The αMSH peptide and the transcript for its precursor, pro-opiomelanocortin (POMC), are similarly present in numerous extra-cutaneous tissues. MC1R genotyping of variants p.(V60M) and p.(R151C) was undertaken for 867 healthy children from the Avon Longitudinal Study of Parent and Children (ALSPAC) cohort, and birthweight modeled using multiple logistic regression analysis. A significant positive association initially found between R151C and birth weight, independent of known birth weight modifiers, was not reproduced when combined with data from an independent genome-wide association study of 6,459 additional members of the same cohort. CONCLUSIONS: These data clearly show a new and hitherto unsuspected role for MC1R in noncutaneous solid tissues before birth.


Asunto(s)
Proteínas Aviares/biosíntesis , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Receptor de Melanocortina Tipo 1/biosíntesis , Animales , Embrión de Pollo , Embrión de Mamíferos/citología , Humanos , Ratones
5.
Genome Res ; 27(8): 1323-1335, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28630177

RESUMEN

While next-generation sequencing has accelerated the discovery of human disease genes, progress has been largely limited to the "low hanging fruit" of mutations with obvious exonic coding or canonical splice site impact. In contrast, the lack of high-throughput, unbiased approaches for functional assessment of most noncoding variants has bottlenecked gene discovery. We report the integration of transcriptome sequencing (RNA-seq), which surveys all mRNAs to reveal functional impacts of variants at the transcription level, into the gene discovery framework for a unique human disease, microcephaly-micromelia syndrome (MMS). MMS is an autosomal recessive condition described thus far in only a single First Nations population and causes intrauterine growth restriction, severe microcephaly, craniofacial anomalies, skeletal dysplasia, and neonatal lethality. Linkage analysis of affected families, including a very large pedigree, identified a single locus on Chromosome 21 linked to the disease (LOD > 9). Comprehensive genome sequencing did not reveal any pathogenic coding or canonical splicing mutations within the linkage region but identified several nonconserved noncoding variants. RNA-seq analysis detected aberrant splicing in DONSON due to one of these noncoding variants, showing a causative role for DONSON disruption in MMS. We show that DONSON is expressed in progenitor cells of embryonic human brain and other proliferating tissues, is co-expressed with components of the DNA replication machinery, and that Donson is essential for early embryonic development in mice as well, suggesting an essential conserved role for DONSON in the cell cycle. Our results demonstrate the utility of integrating transcriptomics into the study of human genetic disease when DNA sequencing alone is not sufficient to reveal the underlying pathogenic mutation.


Asunto(s)
Proteínas de Ciclo Celular/genética , Replicación del ADN , Microcefalia/genética , Microcefalia/patología , Mutación , Proteínas Nucleares/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/patología , Transcriptoma , Animales , Mapeo Cromosómico , Femenino , Ligamiento Genético , Inestabilidad Genómica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Ratones , Ratones Noqueados , Microcefalia/etiología , Osteocondrodisplasias/etiología , Linaje , Embarazo , Empalme del ARN , Análisis de Secuencia de ARN , Secuenciación Completa del Genoma
6.
Wellcome Open Res ; 2: 25, 2017 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-28459107

RESUMEN

BACKGROUND: In humans, the adrenal glands and gonads undergo distinct biological events between 6-10 weeks post conception (wpc), such as testis determination, the onset of steroidogenesis and primordial germ cell development. However, relatively little is currently known about the genetic mechanisms underlying these processes. We therefore aimed to generate a detailed genomic atlas of adrenal and gonad development across these critical stages of human embryonic and fetal development. METHODS: RNA was extracted from 53 tissue samples between 6-10 wpc (adrenal, testis, ovary and control). Affymetrix array analysis was performed and differential gene expression was analysed using Bioconductor. A mathematical model was constructed to investigate time-series changes across the dataset. Pathway analysis was performed using ClueGo and cellular localisation of novel factors confirmed using immunohistochemistry. RESULTS: Using this approach, we have identified novel components of adrenal development (e.g. ASB4, NPR3) and confirmed the role of SRY as the main human testis-determining gene. By mathematical modelling time-series data we have found new genes up-regulated with SOX9 in the testis (e.g. CITED1), which may represent components of the testis development pathway. We have shown that testicular steroidogenesis has a distinct onset at around 8 wpc and identified potential novel components in adrenal and testicular steroidogenesis (e.g. MGARP, FOXO4, MAP3K15, GRAMD1B, RMND2), as well as testis biomarkers (e.g. SCUBE1). We have also shown that the developing human ovary expresses distinct subsets of genes (e.g. OR10G9, OR4D5), but enrichment for established biological pathways is limited. CONCLUSION: This genomic atlas is revealing important novel aspects of human development and new candidate genes for adrenal and reproductive disorders.

7.
Am J Hum Genet ; 99(6): 1338-1352, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27839872

RESUMEN

Anterior segment dysgeneses (ASDs) comprise a spectrum of developmental disorders affecting the anterior segment of the eye. Here, we describe three unrelated families affected by a previously unclassified form of ASD. Shared ocular manifestations include bilateral iris hypoplasia, ectopia lentis, corectopia, ectropion uveae, and cataracts. Whole-exome sequencing and targeted Sanger sequencing identified mutations in CPAMD8 (C3 and PZP-like alpha-2-macroglobulin domain-containing protein 8) as the cause of recessive ASD in all three families. A homozygous missense mutation in the evolutionarily conserved alpha-2-macroglobulin (A2M) domain of CPAMD8, c.4351T>C (p. Ser1451Pro), was identified in family 1. In family 2, compound heterozygous frameshift, c.2352_2353insC (p.Arg785Glnfs∗23), and splice-site, c.4549-1G>A, mutations were identified. Two affected siblings in the third family were compound heterozygous for splice-site mutations c.700+1G>T and c.4002+1G>A. CPAMD8 splice-site mutations caused aberrant pre-mRNA splicing in vivo or in vitro. Intriguingly, our phylogenetic analysis revealed rodent lineage-specific CPAMD8 deletion, precluding a developmental expression study in mice. We therefore investigated the spatiotemporal expression of CPAMD8 in the developing human eye. RT-PCR and in situ hybridization revealed CPAMD8 expression in the lens, iris, cornea, and retina early in development, including strong expression in the distal tips of the retinal neuroepithelium that form the iris and ciliary body, thus correlating CPAMD8 expression with the affected tissues. Our study delineates a unique form of recessive ASD and defines a role for CPAMD8, a protein of unknown function, in anterior segment development, implying another pathway for the pathogenicity of ASD.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Complemento C3/genética , Anomalías del Ojo/genética , Genes Recesivos/genética , Mutación , Inhibidor de Tripsina Pancreática de Kazal/genética , alfa-Macroglobulinas/genética , Adolescente , Adulto , Secuencia de Aminoácidos , Segmento Anterior del Ojo/metabolismo , Niño , Preescolar , Complemento C3/química , Femenino , Humanos , Masculino , Persona de Mediana Edad , Inhibidor de Tripsina Pancreática de Kazal/química , Adulto Joven , alfa-Macroglobulinas/química
9.
J Anat ; 228(3): 452-63, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26612825

RESUMEN

TOP2A and TOP2B are type II topoisomerase enzymes that have important but distinct roles in DNA replication and RNA transcription. Recently, TOP2B has been implicated in the transcription of long genes in particular that play crucial roles in neural development and are susceptible to mutations contributing to neurodevelopmental conditions such as autism and schizophrenia. This study maps their expression in the early foetal human telencephalon between 9 and 12 post-conceptional weeks. TOP2A immunoreactivity was restricted to cell nuclei of the proliferative layers of the cortex and ganglionic eminences (GE), including the ventricular zone and subventricular zone (SVZ) closely matching expression of the proliferation marker KI67. Comparison with sections immunolabelled for NKX2.1, a medial GE (MGE) marker, and PAX6, a cortical progenitor cell and lateral GE (LGE) marker, revealed that TOP2A-expressing cells were more abundant in MGE than the LGE. In the cortex, TOP2B is expressed in cell nuclei in both proliferative (SVZ) and post-mitotic compartments (intermediate zone and cortical plate) as revealed by comparison with immunostaining for PAX6 and the post-mitotic neuron marker TBR1. However, co-expression with KI67 was rare. In the GE, TOP2B was also expressed by proliferative and post-mitotic compartments. In situ hybridisation studies confirmed these patterns of expression, except that TOP2A mRNA is restricted to cells in the G2/M phase of division. Thus, during early development, TOP2A is likely to have a role in cell proliferation, whereas TOP2B is expressed in post-mitotic cells and may be important in controlling expression of long genes even at this early stage.


Asunto(s)
Antígenos de Neoplasias/biosíntesis , ADN-Topoisomerasas de Tipo II/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Feto/enzimología , Neurogénesis/fisiología , Telencéfalo/embriología , Telencéfalo/enzimología , Humanos , Inmunohistoquímica , Hibridación in Situ , Proteínas de Unión a Poli-ADP-Ribosa
10.
Development ; 142(18): 3073-6, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26395135

RESUMEN

Congenital anomalies are a significant burden on human health. Understanding the developmental origins of such anomalies is key to developing potential therapies. The Human Developmental Biology Resource (HDBR), based in London and Newcastle, UK, was established to provide embryonic and fetal material for a variety of human studies ranging from single gene expression analysis to large-scale genomic/transcriptomic studies. Increasingly, HDBR material is enabling the derivation of stem cell lines and contributing towards developments in tissue engineering. Use of the HDBR and other fetal tissue resources discussed here will contribute to the long-term aims of understanding the causation and pathogenesis of congenital anomalies, and developing new methods for their treatment and prevention.


Asunto(s)
Anomalías Congénitas/fisiopatología , Investigaciones con Embriones , Investigación Fetal , Investigación con Células Madre , Bancos de Tejidos/tendencias , Ingeniería de Tejidos/tendencias , Anomalías Congénitas/diagnóstico , Inglaterra , Humanos , Ingeniería de Tejidos/métodos
11.
Mol Cell Endocrinol ; 351(2): 264-8, 2012 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-22240064

RESUMEN

Lin28 proteins are emerging as important regulators of microRNAs in endocrine systems. Lin28a regulates primordial germ cell development and puberty timing in mice, whereas the related protein LIN28B is associated with age at menarche in genome-wide association studies in humans. Here, we studied expression of LIN28A and LIN28B in early human gonad development. LIN28A increased in the developing ovary between 6 and 9weeks post conception, but not in the developing testis. Immunohistochemistry demonstrated LIN28A in peripheral germ cells. LIN28B was expressed at lower levels in both tissues and did not increase with time. As disruption of Lin28a affects germ cell development in mice, LIN28A was considered a candidate gene for primary ovarian insufficiency (POI) in humans. However, no significant changes were found in 50 women studied. These findings show LIN28A is strongly expressed in germ cells during early human ovary development, but disruption of LIN28A is not a common cause of POI.


Asunto(s)
Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ovario/embriología , Insuficiencia Ovárica Primaria/genética , Animales , Secuencia de Bases , Análisis Mutacional de ADN , Femenino , Células Germinativas/metabolismo , Humanos , Masculino , Ratones , MicroARNs/biosíntesis , MicroARNs/genética , MicroARNs/metabolismo , Ovario/metabolismo , Insuficiencia Ovárica Primaria/metabolismo , Insuficiencia Ovárica Primaria/patología , Proteínas de Unión al ARN , Análisis de Secuencia de ADN , Testículo/embriología , Testículo/metabolismo
12.
Invest Ophthalmol Vis Sci ; 53(1): 440-7, 2012 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-22110064

RESUMEN

PURPOSE: To identify heritable genetic factors altering susceptibility to refractive error in the general population. METHODS: This was a genetic association study of refractive error investigating genetic polymorphisms in regions previously reported through linkage. Two study panels were drawn from the British 1958 Birth Cohort, composed of 2211 persons 44 years of age at the time of visit. Two main outcomes were considered: refractive error as a continuous outcome (spherical equivalent) and myopia as a diagnosis (defined as spherical equivalent equal to or worse than-1.00 diopter). Genotyping was initially performed in 1188 subjects from the outer tertiles of the population distribution, using customized arrays of single nucleotide polymorphisms (SNPs) saturating regions of previously reported highly significant linkage. In a second stage, SNPs most significantly associated were validated in 1023 more persons. Findings were investigated further through human fetal expression studies. RESULTS: Polymorphisms within the SERPINI2 gene were associated with refractive error in two different European subgroups from the 1958 British Birth Cohort (meta-analysis P = 7.4E-05 for rs9810473). Association was also significant for myopia (best association: OR = 0.80; 95% CI, 0.69-0.93; P = 0.003 for rs10936538). Expression profiling of SERPINI2 revealed that the gene is expressed in the retina and in other eye and CNS tissues. CONCLUSIONS: The novel association of SERPINI2 with refractive error and myopia is suggestive of a possible link between physiological pathways controlling eye growth and development and those controlling glucose metabolism. The findings indicate that SERPINI2 is a promising candidate for further investigations of the genetic susceptibility to myopia.


Asunto(s)
Predisposición Genética a la Enfermedad , Miopía/genética , Proteínas de Neoplasias/genética , Polimorfismo de Nucleótido Simple , Serpinas/genética , Adulto , Encéfalo/embriología , Estudios de Cohortes , Ojo/embriología , Feto/metabolismo , Perfilación de la Expresión Génica , Genotipo , Técnicas de Genotipaje , Humanos , Hibridación in Situ , Persona de Mediana Edad , Proteínas de Neoplasias/metabolismo , Serpinas/metabolismo , Reino Unido
13.
J Clin Endocrinol Metab ; 96(10): E1709-18, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21832120

RESUMEN

CONTEXT: Fibroblast growth factor (FGF) 8 is important for GnRH neuronal development with human mutations resulting in Kallmann syndrome. Murine data suggest a role for Fgf8 in hypothalamo-pituitary development; however, its role in the etiology of wider hypothalamo-pituitary dysfunction in humans is unknown. OBJECTIVE: The objective of this study was to screen for FGF8 mutations in patients with septo-optic dysplasia (n = 374) or holoprosencephaly (HPE)/midline clefts (n = 47). METHODS: FGF8 was analyzed by PCR and direct sequencing. Ethnically matched controls were then screened for mutated alleles (n = 480-686). Localization of Fgf8/FGF8 expression was analyzed by in situ hybridization in developing murine and human embryos. Finally, Fgf8 hypomorphic mice (Fgf8(loxPNeo/-)) were analyzed for the presence of forebrain and hypothalamo-pituitary defects. RESULTS: A homozygous p.R189H mutation was identified in a female patient of consanguineous parentage with semilobar HPE, diabetes insipidus, and TSH and ACTH insufficiency. Second, a heterozygous p.Q216E mutation was identified in a female patient with an absent corpus callosum, hypoplastic optic nerves, and Moebius syndrome. FGF8 was expressed in the ventral diencephalon and anterior commissural plate but not in Rathke's pouch, strongly suggesting early onset hypothalamic and corpus callosal defects in these patients. This was consolidated by significantly reduced vasopressin and oxytocin staining neurons in the hypothalamus of Fgf8 hypomorphic mice compared with controls along with variable hypothalamo-pituitary defects and HPE. CONCLUSION: We implicate FGF8 in the etiology of recessive HPE and potentially septo-optic dysplasia/Moebius syndrome for the first time to our knowledge. Furthermore, FGF8 is important for the development of the ventral diencephalon, hypothalamus, and pituitary.


Asunto(s)
Anomalías Craneofaciales/genética , Factor 8 de Crecimiento de Fibroblastos/genética , Holoprosencefalia/genética , Enfermedades Hipotalámicas/genética , Sistema Hipotálamo-Hipofisario/fisiopatología , Mutación/fisiología , Enfermedades de la Hipófisis/genética , Agenesia del Cuerpo Calloso/complicaciones , Agenesia del Cuerpo Calloso/genética , Arginina Vasopresina/metabolismo , Análisis Mutacional de ADN , Femenino , Factor 8 de Crecimiento de Fibroblastos/fisiología , Hormona de Crecimiento Humana/sangre , Humanos , Hidrocortisona/sangre , Inmunohistoquímica , Hibridación in Situ , Lactante , Imagen por Resonancia Magnética , Hipófisis/crecimiento & desarrollo , Hipófisis/fisiología , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/biosíntesis , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Displasia Septo-Óptica/genética , Tirotropina/sangre
14.
PLoS One ; 6(1): e16366, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21297984

RESUMEN

Human testis development starts from around 42 days post conception with a transient wave of SRY expression followed by up-regulation of testis specific genes and a distinct set of morphological, paracrine and endocrine events. Although anatomical changes in the ovary are less marked, a distinct sub-set of ovary specific genes are also expressed during this time. The furin-domain containing peptide R-spondin1 (RSPO1) has recently emerged as an important regulator of ovary development through up-regulation of the WNT/ß-catenin pathway to oppose testis formation. Here, we show that RSPO1 is upregulated in the ovary but not in the testis during critical early stages of gonad development in humans (between 6-9 weeks post conception), whereas the expression of the related genes WNT4 and CTNNB1 (encoding ß catenin) is not significantly different between these tissues. Furthermore, reduced R-spondin1 function in the ovotestis of an individual (46,XX) with a RSPO1 mutation leads to reduced ß-catenin protein and WNT4 mRNA levels, consistent with down regulation of ovarian pathways. Transfection of wild-type RSPO1 cDNA resulted in weak dose-dependent activation of a ß-catenin responsive TOPFLASH reporter (1.8 fold maximum), whereas co-transfection of CTNNB1 (encoding ß-catenin) with RSPO1 resulted in dose-dependent synergistic augmentation of this reporter (approximately 10 fold). Furthermore, R-spondin1 showed strong nuclear localization in several different cell lines. Taken together, these data show that R-spondin1 is upregulated during critical stages of early human ovary development and may function as a tissue-specific amplifier of ß-catenin signaling to oppose testis determination.


Asunto(s)
Ovario/crecimiento & desarrollo , Transducción de Señal , Trombospondinas/genética , beta Catenina/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Ovario/embriología , ARN Mensajero , Testículo/embriología , Testículo/crecimiento & desarrollo , Proteínas Wnt/genética , Proteína Wnt4 , beta Catenina/genética
15.
J Med Genet ; 48(1): 48-54, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20972252

RESUMEN

BACKGROUND: Autism spectrum disorder (ASD) is characterised by impairments in social communication and by a pattern of repetitive behaviours, with learning disability (LD) typically seen in up to 70% of cases. A recent study using the PPL statistical framework identified a novel region of genetic linkage on chromosome 16q21 that is limited to ASD families with LD. METHODS: In this study, two families with autism and/or LD are described which harbour rare >1.6 Mb microdeletions located within this linkage region. The deletion breakpoints are mapped at base-pair resolution and segregation analysis is performed using a combination of 1M single nucleotide polymorphism (SNP) technology, array comparative genomic hybridisation (CGH), long-range PCR, and Sanger sequencing. The frequency of similar genomic variants in control subjects is determined through analysis of published SNP array data. Expression of CDH8, the only gene disrupted by these microdeletions, is assessed using reverse transcriptase PCR and in situ hybridisation analysis of 9 week human embryos. RESULTS: The deletion of chr16: 60 025 584-61 667 839 was transmitted to three of three boys with autism and LD and none of four unaffected siblings, from their unaffected mother. In a second family, an overlapping deletion of chr16: 58 724 527-60 547 472 was transmitted to an individual with severe LD from his father with moderate LD. No copy number variations (CNVs) disrupting CDH8 were observed in 5023 controls. Expression analysis indicates that the two CDH8 isoforms are present in the developing human cortex. CONCLUSION: Rare familial 16q21 microdeletions and expression analysis implicate CDH8 in susceptibility to autism and LD.


Asunto(s)
Trastorno Autístico/genética , Cadherinas/genética , Deleción Cromosómica , Cromosomas Humanos Par 16/genética , Ligamiento Genético , Predisposición Genética a la Enfermedad , Discapacidades para el Aprendizaje/genética , Adolescente , Secuencia de Bases , Cadherinas/metabolismo , Niño , Variaciones en el Número de Copia de ADN/genética , Análisis Mutacional de ADN , Familia , Femenino , Regulación de la Expresión Génica , Genoma Humano/genética , Humanos , Pruebas de Inteligencia , Internet , Masculino , Datos de Secuencia Molecular , Linaje , Adulto Joven
16.
J Anat ; 216(6): 683-91, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20408909

RESUMEN

In Drosophila, Iroquois (Irx) genes have various functions including the specification of the identity of wing veins. Vertebrate Iroquois (Irx) genes have been reported to be expressed in the developing digits of mouse limbs. Here we carry out a phylogenetic analysis of vertebrate Irx genes and compare expression in developing limbs of mouse, chick and human embryos and in zebrafish pectoral fin buds. We confirm that the six Irx gene families in vertebrates are well defined and that Clusters A and B are duplicates; in contrast, Irx1 and 3, Irx2 and 5, and Irx4 and 6 are paralogs. All Irx genes in mouse and chick are expressed in developing limbs. Detailed comparison of the expression patterns in mouse and chick shows that expression patterns of genes in the same cluster are generally similar but paralogous genes have different expression patterns. Mouse and chick Irx1 are expressed in digit condensations, whereas mouse and chick Irx6 are expressed interdigitally. The timing of Irx1 expression in individual digits in mouse and chick is different. Irx1 is also expressed in digit condensations in developing human limbs, thus showing conservation of expression of this gene in higher vertebrates. In zebrafish, Irx genes of all but six of the families are expressed in early stage pectoral fin buds but not at later stages, suggesting that these genes are not involved in patterning distal structures in zebrafish fins.


Asunto(s)
Desarrollo Embrionario/genética , Proteínas de Homeodominio/genética , Filogenia , Pez Cebra/genética , Animales , Embrión de Pollo , Extremidades/embriología , Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Vertebrados/genética , Pez Cebra/embriología
17.
Hum Genet ; 126(6): 791-803, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19685247

RESUMEN

Mutations in the transcription factor encoding TFAP2A gene underlie branchio-oculo-facial syndrome (BOFS), a rare dominant disorder characterized by distinctive craniofacial, ocular, ectodermal and renal anomalies. To elucidate the range of ocular phenotypes caused by mutations in TFAP2A, we took three approaches. First, we screened a cohort of 37 highly selected individuals with severe ocular anomalies plus variable defects associated with BOFS for mutations or deletions in TFAP2A. We identified one individual with a de novo TFAP2A four amino acid deletion, a second individual with two non-synonymous variations in an alternative splice isoform TFAP2A2, and a sibling-pair with a paternally inherited whole gene deletion with variable phenotypic expression. Second, we determined that TFAP2A is expressed in the lens, neural retina, nasal process, and epithelial lining of the oral cavity and palatal shelves of human and mouse embryos--sites consistent with the phenotype observed in patients with BOFS. Third, we used zebrafish to examine how partial abrogation of the fish ortholog of TFAP2A affects the penetrance and expressivity of ocular phenotypes due to mutations in genes encoding bmp4 or tcf7l1a. In both cases, we observed synthetic, enhanced ocular phenotypes including coloboma and anophthalmia when tfap2a is knocked down in embryos with bmp4 or tcf7l1a mutations. These results reveal that mutations in TFAP2A are associated with a wide range of eye phenotypes and that hypomorphic tfap2a mutations can increase the risk of developmental defects arising from mutations at other loci.


Asunto(s)
Anomalías del Ojo/genética , Ojo/embriología , Retina/anomalías , Factor de Transcripción AP-2/genética , Adulto , Animales , Síndrome Branquio Oto Renal/genética , Preescolar , Femenino , Eliminación de Gen , Humanos , Lactante , Masculino , Persona de Mediana Edad , Morfogénesis/genética , Mutación , Pez Cebra , Proteínas de Pez Cebra/genética
18.
Am J Hum Genet ; 84(5): 698-705, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19409524

RESUMEN

We describe a recessively inherited frontonasal malformation characterized by a distinctive facial appearance, with hypertelorism, wide nasal bridge, short nasal ridge, bifid nasal tip, broad columella, widely separated slit-like nares, long philtrum with prominent bilateral swellings, and midline notch in the upper lip and alveolus. Additional recurrent features present in a minority of individuals have been upper eyelid ptosis and midline dermoid cysts of craniofacial structures. Assuming recessive inheritance, we mapped the locus in three families to chromosome 1 and identified mutations in ALX3, which is located at band 1p13.3 and encodes the aristaless-related ALX homeobox 3 transcription factor. In total, we identified seven different homozygous pathogenic mutations in seven families. These mutations comprise missense substitutions at critical positions within the conserved homeodomain as well as nonsense, frameshift, and splice-site mutations, all predicting severe or complete loss of function. Our findings contrast with previous studies of the orthologous murine gene, which showed no phenotype in Alx3(-/-) homozygotes, apparently as a result of functional redundancy with the paralogous Alx4 gene. We conclude that ALX3 is essential for normal facial development in humans and that deficiency causes a clinically recognizable phenotype, which we term frontorhiny.


Asunto(s)
Anomalías Craneofaciales/genética , Proteínas de Homeodominio/genética , Hueso Nasal/anomalías , Niño , Cromosomas Humanos Par 1/genética , Humanos , Recién Nacido , Mutación
19.
J Clin Endocrinol Metab ; 94(2): 678-83, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18984668

RESUMEN

CONTEXT: Disorders of adrenal development result in significant morbidity and mortality. However, the molecular basis of human adrenal development, and many forms of disease, is still poorly understood. OBJECTIVES: We evaluated the role of two new candidate genes, CBP/p300-interacting transactivator, with Glu/Asp-rich C-terminal domain, 2 (CITED2), and pre-B-cell leukemia transcription factor 1 (PBX1), in human adrenal development and disease. DESIGN: CITED2 and PBX1 expression in early human fetal adrenal development was assessed using RT-PCR and in situ hybridization. The regulation of CITED2 and PBX1 by steroidogenic factor-1 (SF-1) and dosage-sensitive sex reversal, adrenal hypoplasia congenital, critical region on the X chromosome, gene-1 (DAX1) was evaluated in NCI-H295R human adrenocortical tumor cells by studying promoter regulation. Finally, mutational analysis of CITED2 and PBX1 was performed in patients with primary adrenal disorders. RESULTS: CITED2 and PBX1 are expressed in the human fetal adrenal gland during early development. Both genes are activated by SF-1 in a dose-dependent manner in NCI-H295R cells, and, surprisingly, PBX1 is synergistically activated by SF-1 and DAX1. Mutational analysis failed to reveal significant coding sequence changes in individuals with primary adrenal disorders. CONCLUSIONS: CITED2 and PBX1 are likely to be important mediators of adrenal development and function in humans, but mutations in these genes are not common causes of adrenal failure in patients in whom a molecular diagnosis remains unknown. The positive interaction between DAX1 and SF-1 in regulating PBX1 may be an important mechanism in this process.


Asunto(s)
Enfermedades de las Glándulas Suprarrenales/genética , Glándulas Suprarrenales/embriología , Proteínas de Unión al ADN/fisiología , Proteínas Proto-Oncogénicas/fisiología , Proteínas Represoras/fisiología , Transactivadores/fisiología , Enfermedades de las Glándulas Suprarrenales/patología , Glándulas Suprarrenales/metabolismo , Glándulas Suprarrenales/patología , Células Cultivadas , Receptor Nuclear Huérfano DAX-1 , Análisis Mutacional de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Mutación/fisiología , Factor de Transcripción 1 de la Leucemia de Células Pre-B , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Receptores de Ácido Retinoico/metabolismo , Receptores de Ácido Retinoico/fisiología , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Factor Esteroidogénico 1/metabolismo , Factor Esteroidogénico 1/fisiología , Transactivadores/genética , Transactivadores/metabolismo , Transfección
20.
Dis Model Mech ; 1(4-5): 241-54, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-19093031

RESUMEN

A homozygous substitution of the highly conserved isoleucine at position 26 by threonine (I26T) in the transcriptional repressor HESX1 has been associated with anterior pituitary hypoplasia in a human patient, with no forebrain or eye defects. Two individuals carrying a homozygous substitution of the conserved arginine at position 160 by cysteine (R160C) manifest septo-optic dysplasia (SOD), a condition characterised by pituitary abnormalities associated with midline telencephalic structure defects and optic nerve hypoplasia. We have generated two knock-in mouse models containing either the I26T or R160C substitution in the genomic locus. Hesx1(I26T/I26T) embryos show pituitary defects comparable with Hesx1(-/-) mouse mutants, with frequent occurrence of ocular abnormalities, although the telencephalon develops normally. Hesx1(R160C/R160C) mutants display forebrain and pituitary defects that are identical to those observed in Hesx1(-/-) null mice. We also show that the expression pattern of HESX1 during early human development is very similar to that described in the mouse, suggesting that the function of HESX1 is conserved between the two species. Together, these results suggest that the I26T mutation yields a hypomorphic allele, whereas R160C produces a null allele and, consequently, a more severe phenotype in both mice and humans.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de Homeodominio/genética , Hipopituitarismo/genética , Mutación , Proteínas Represoras/genética , Displasia Septo-Óptica/genética , Animales , Tipificación del Cuerpo , Ratones , Prosencéfalo/anomalías , Prosencéfalo/embriología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...