Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 106(2): 143-152, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32032513

RESUMEN

Advances in genomics have transformed our ability to identify the genetic causes of rare diseases (RDs), yet we have a limited understanding of the mechanistic roles of most genes in health and disease. When a novel RD gene is first discovered, there is minimal insight into its biological function, the pathogenic mechanisms of disease-causing variants, and how therapy might be approached. To address this gap, the Canadian Rare Diseases Models and Mechanisms (RDMM) Network was established to connect clinicians discovering new disease genes with Canadian scientists able to study equivalent genes and pathways in model organisms (MOs). The Network is built around a registry of more than 500 Canadian MO scientists, representing expertise for over 7,500 human genes. RDMM uses a committee process to identify and evaluate clinician-MO scientist collaborations and approve 25,000 Canadian dollars in catalyst funding. To date, we have made 85 clinician-MO scientist connections and funded 105 projects. These collaborations help confirm variant pathogenicity and unravel the molecular mechanisms of RD, and also test novel therapies and lead to long-term collaborations. To expand the impact and reach of this model, we made the RDMM Registry open-source, portable, and customizable, and we freely share our committee structures and processes. We are currently working with emerging networks in Europe, Australia, and Japan to link international RDMM networks and registries and enable matches across borders. We will continue to create meaningful collaborations, generate knowledge, and advance RD research locally and globally for the benefit of patients and families living with RD.


Asunto(s)
Modelos Animales de Enfermedad , Marcadores Genéticos , Enfermedades Raras/genética , Enfermedades Raras/terapia , Sistema de Registros/normas , Animales , Bases de Datos Factuales , Genómica , Humanos , Enfermedades Raras/epidemiología
2.
Genesis ; 57(1): e23278, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30614633

RESUMEN

Fetal Alcohol Spectrum Disorder (FASD) is a set of neurodevelopmental malformations caused by maternal consumption of alcohol during pregnancy. FASD sentinel facial features are unique to the disorder, and microcephaly is common in severe forms of FASD. Retinoic acid deficiency has been shown to cause craniofacial malformations and microcephaly in animal models reminiscent of those caused by prenatal alcohol exposure. Alcohol exposure affects the migration and survival of cranial neural crest cells, which are required for proper frontonasal prominence and pharyngeal arch development. Defects in craniofacial development are further amplified by the many downstream pathways that are transcriptionally controlled retinoic acid target genes, including Shh signaling. Recent evidence shows that alcohol exposure itself is sufficient to induce retinoic acid deficiency in the embryo. These data suggest that retinoic acid deficiency is an important underlying etiology of FASD. In disorders like Vitamin A Deficiency, FASD, DiGeorge (22q11.2 Deletion Syndrome), CHARGE, Smith-Magenis, Matthew-Wood, and Congenital Zika Syndromes, evidence is accumulating to link reduced retinoic acid signaling with developmental defects like craniofacial malformations and microcephaly. Research focus on characterizing the effects of retinoic acid deficiency during early development and on understanding the downstream signaling pathways involved in aberrant head, and craniofacial development will reveal underlying etiologies of these disorders.


Asunto(s)
Anomalías Craneofaciales/etiología , Trastornos del Espectro Alcohólico Fetal/etiología , Microcefalia/etiología , Cresta Neural/embriología , Tretinoina/metabolismo , Animales , Humanos , Cresta Neural/metabolismo
3.
Biochem Cell Biol ; 96(2): 131-147, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29370535

RESUMEN

The potential impact of prenatal alcohol exposure (PAE) varies considerably among exposed individuals, with some displaying serious alcohol-related effects and many others showing few or no overt signs of fetal alcohol spectrum disorder (FASD). In animal models, variables such as nutrition, genetic background, health, other drugs, and stress, as well as dosage, duration, and gestational timing of exposure to alcohol can all be controlled in a way that is not possible in a clinical situation. In this review we examine mouse models of PAE and focus on those with demonstrated craniofacial malformations, abnormal brain development, or behavioral phenotypes that may be considered FASD-like outcomes. Analysis of these data should provide a valuable tool for researchers wishing to choose the PAE model best suited to their research questions or to investigate established PAE models for FASD comorbidities. It should also allow recognition of patterns linking gestational timing, dosage, and duration of PAE, such as recognizing that binge alcohol exposure(s) during early gestation can lead to severe FASD outcomes. Identified patterns could be particularly insightful and lead to a better understanding of the molecular mechanisms underlying FASD.


Asunto(s)
Consumo Excesivo de Bebidas Alcohólicas/metabolismo , Modelos Animales de Enfermedad , Trastornos del Espectro Alcohólico Fetal/metabolismo , Efectos Tardíos de la Exposición Prenatal/metabolismo , Animales , Consumo Excesivo de Bebidas Alcohólicas/complicaciones , Consumo Excesivo de Bebidas Alcohólicas/patología , Femenino , Trastornos del Espectro Alcohólico Fetal/patología , Humanos , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/patología
4.
Biochem Cell Biol ; 96(2): 275-287, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29544064

RESUMEN

Translating to the Community (T2C) is a social biorepository designed to advance new diagnostic tools and realign community-clinical processes, with the aim to mitigate the short- and long-term impacts of fetal alcohol spectrum disorder (FASD) as well as prenatal alcohol exposure and its co-morbidities and behaviors. In this paper, we describe the evolution of this repository as a new translational partnership to advance a precision-medicine approach to FASD. Key to its evolution was a partnership between academic researchers, Indigenous communities, families, and a regional diagnostic clinic. We further describe the rationale for social biobanking, the type of banking, ethical engagement of families, communities, and clinics, their roles in repository design, governance, translation, and research activities, types of data collected from families, and how the study data are managed, reported, and accessed. The repository design includes biological samples, social-contextual health-survey data, and clinical data (which are linkable to administrative data) from community and clinical cohorts of diagnosed children, children prenatally exposed but not diagnosed, children suspected to have had a prenatal exposure, and related siblings, biological parents, and unrelated children and their parents. From these cohorts and families, potential studies drawing on this data will shed light on various risk factors, social and biological pathways, and service utilization issues, with the aim to implement primary and secondary prevention and intervention strategies.


Asunto(s)
Bancos de Muestras Biológicas , Epigénesis Genética , Trastornos del Espectro Alcohólico Fetal , Adolescente , Bancos de Muestras Biológicas/ética , Bancos de Muestras Biológicas/organización & administración , Bancos de Muestras Biológicas/normas , Canadá , Niño , Preescolar , Femenino , Humanos , Masculino
5.
Biochem Cell Biol ; 96(2): 161-166, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29533680

RESUMEN

Fetal alcohol spectrum disorder (FASD) is characterized by a combination of neurological, developmental, and congenital defects that may occur as a consequence of prenatal alcohol exposure. Earlier reports showed that large chromosomal anomalies may link to FASD. Here, we examined the prevalence and types of copy number variations (CNVs) in FASD cases previously diagnosed by a multidisciplinary FASD team in sites across Canada. We genotyped 95 children with FASD and 87 age-matched, typically developing controls on the Illumina Human Omni2.5 SNP (single nucleotide polymorphisms) array platform. We compared their CNVs with those of 10 851 population controls to identify rare CNVs (<0.1% frequency), which may include large unbalanced chromosomal abnormalities, that might be relevant to FASD. In 12/95 (13%) of the FASD cases, rare CNVs were found that impact potentially clinically relevant developmental genes, including the CACNA1H involved in epilepsy and autism, the 3q29 deletion disorder, and others. Our results show that a subset of children diagnosed with FASD have chromosomal deletions and duplications that may co-occur or explain the neurodevelopmental impairments in a diagnosed cohort of FASD individuals. Children suspected to have FASD with or without sentinel facial features of fetal alcohol syndrome and neurodevelopmental delays should potentially be evaluated by a clinical geneticist and possibly have genetic investigations as appropriate to exclude other etiologies.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 3/genética , Trastornos del Espectro Alcohólico Fetal/genética , Dosificación de Gen , Polimorfismo de Nucleótido Simple , Niño , Preescolar , Femenino , Humanos , Masculino
6.
PLoS Genet ; 9(10): e1003895, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-24204307

RESUMEN

The gene encoding a DNA/RNA binding protein FUS/TLS is frequently mutated in amyotrophic lateral sclerosis (ALS). Mutations commonly affect its carboxy-terminal nuclear localization signal, resulting in varying deficiencies of FUS nuclear localization and abnormal cytoplasmic accumulation. Increasing evidence suggests deficiencies in FUS nuclear function may contribute to neuron degeneration. Here we report a novel FUS autoregulatory mechanism and its deficiency in ALS-associated mutants. Using FUS CLIP-seq, we identified significant FUS binding to a highly conserved region of exon 7 and the flanking introns of its own pre-mRNAs. We demonstrated that FUS is a repressor of exon 7 splicing and that the exon 7-skipped splice variant is subject to nonsense-mediated decay (NMD). Overexpression of FUS led to the repression of exon 7 splicing and a reduction of endogenous FUS protein. Conversely, the repression of exon 7 was reduced by knockdown of FUS protein, and moreover, it was rescued by expression of EGFP-FUS. This dynamic regulation of alternative splicing describes a novel mechanism of FUS autoregulation. Given that ALS-associated FUS mutants are deficient in nuclear localization, we examined whether cells expressing these mutants would be deficient in repressing exon 7 splicing. We showed that FUS harbouring R521G, R522G or ΔExon15 mutation (minor, moderate or severe cytoplasmic localization, respectively) directly correlated with respectively increasing deficiencies in both exon 7 repression and autoregulation of its own protein levels. These data suggest that compromised FUS autoregulation can directly exacerbate the pathogenic accumulation of cytoplasmic FUS protein in ALS. We showed that exon 7 skipping can be induced by antisense oligonucleotides targeting its flanking splice sites, indicating the potential to alleviate abnormal cytoplasmic FUS accumulation in ALS. Taken together, FUS autoregulation by alternative splicing provides insight into a molecular mechanism by which FUS-regulated pre-mRNA processing can impact a significant number of targets important to neurodegeneration.


Asunto(s)
Empalme Alternativo/genética , Esclerosis Amiotrófica Lateral/genética , Regulación de la Expresión Génica/genética , Proteína FUS de Unión a ARN , Esclerosis Amiotrófica Lateral/etiología , Esclerosis Amiotrófica Lateral/patología , Citoplasma/genética , Exones/genética , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Células HeLa , Humanos , Intrones/genética , Mutación , Precursores del ARN/biosíntesis , Precursores del ARN/genética , Proteína FUS de Unión a ARN/biosíntesis , Proteína FUS de Unión a ARN/genética
7.
Biomolecules ; 14(5)2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38785976

RESUMEN

Fetal Alcohol Spectrum Disorder (FASD) is a common neurodevelopmental disorder that affects an estimated 2-5% of North Americans. FASD is induced by prenatal alcohol exposure (PAE) during pregnancy and while there is a clear genetic contribution, few genetic factors are currently identified or understood. In this study, using a candidate gene approach, we performed a genetic variant analysis of retinoic acid (RA) metabolic and developmental signaling pathway genes on whole exome sequencing data of 23 FASD-diagnosed individuals. We found risk and resilience alleles in ADH and ALDH genes known to normally be involved in alcohol detoxification at the expense of RA production, causing RA deficiency, following PAE. Risk and resilience variants were also identified in RA-regulated developmental pathway genes, especially in SHH and WNT pathways. Notably, we also identified significant variants in the causative genes of rare neurodevelopmental disorders sharing comorbidities with FASD, including STRA6 (Matthew-Wood), SOX9 (Campomelic Dysplasia), FDG1 (Aarskog), and 22q11.2 deletion syndrome (TBX1). Although this is a small exploratory study, the findings support PAE-induced RA deficiency as a major etiology underlying FASD and suggest risk and resilience variants may be suitable biomarkers to determine the risk of FASD outcomes following PAE.


Asunto(s)
Trastornos del Espectro Alcohólico Fetal , Tretinoina , Humanos , Femenino , Tretinoina/metabolismo , Trastornos del Espectro Alcohólico Fetal/genética , Trastornos del Espectro Alcohólico Fetal/metabolismo , Embarazo , Masculino , Predisposición Genética a la Enfermedad , Secuenciación del Exoma
8.
Mamm Genome ; 23(9-10): 580-6, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22968824

RESUMEN

In 2007, the International Knockout Mouse Consortium (IKMC) made the ambitious promise to generate mutations in virtually every protein-coding gene of the mouse genome in a concerted worldwide action. Now, 5 years later, the IKMC members have developed high-throughput gene trapping and, in particular, gene-targeting pipelines and generated more than 17,400 mutant murine embryonic stem (ES) cell clones and more than 1,700 mutant mouse strains, most of them conditional. A common IKMC web portal (www.knockoutmouse.org) has been established, allowing easy access to this unparalleled biological resource. The IKMC materials considerably enhance functional gene annotation of the mammalian genome and will have a major impact on future biomedical research.


Asunto(s)
Ratones Noqueados/genética , Animales , Internacionalidad , Internet , Ratones
9.
Pharmacoecon Open ; 6(2): 253-263, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34628597

RESUMEN

BACKGROUND AND AIMS: Fetal alcohol spectrum disorder (FASD) is a condition that results from prenatal alcohol exposure. Though diagnosis is important for individuals with FASD to receive appropriate care, diagnosis can be difficult to obtain. Accurate diagnoses can be impeded because of an inability to confirm prenatal alcohol exposure. This is particularly problematic in instances when family cannot confirm prenatal alcohol exposure. DNA methylation testing represents a novel approach to identifying prenatal alcohol exposure via epigenetic biomarkers. The objective was to assess the impact on laboratory expenditures from adopting DNA methylation additively to the diagnostic workup for patients suspected of having FASD for whom prenatal alcohol exposure cannot be otherwise confirmed. METHODS: A budget impact model was developed that incorporates laboratory cost data, population data for Manitoba Canada, literature, and expert opinion. Probabilistic analysis was conducted for the primary analysis and deterministic sensitivity analyses were conducted to assess the sensitivity of the budget impact to changes in model parameters. The perspective of the present study is that of the laboratory budget holder at a centralized laboratory in Manitoba, Canada. RESULTS: Over a 5-year period, it was estimated that there would be 500 DNA methylation tests and the predicted budget impact to the laboratory budget holder was $207,574 (95% credible interval: 70,208-408,161) in Canadian dollars (CAD). Over a 10-year period, it was estimated that there would be 1017 DNA methylation tests and the predicted budget impact to the laboratory budget holder was CAD$439,470 (95% credible interval: 148,902-867,328). CONCLUSIONS: Findings provide insight into the impact that DNA methylation testing would have on laboratory budgets if used in the diagnostic workup for FASD in individuals for whom prenatal alcohol exposure cannot be confirmed otherwise.

10.
BMC Biol ; 8: 17, 2010 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-20199662

RESUMEN

BACKGROUND: The Dlc1 (deleted in liver cancer 1) tumour suppressor gene codes for a RhoGTPase activating protein that is found inactivated in many tumour types. Several transcriptional isoforms have been described but the functional significance and tissue distribution of each form is presently poorly understood. Also, differences in the number of isoforms and splice variants reported still exist between different mammalian species. In order to better understand the number and function of the different variants of the Dlc1 gene in the mouse, we have carried out a detailed analysis. Extensive 3' RACE experiments were carried out in order to identify all possible Dlc1 isoforms and splice variants in the mouse. In addition, we have generated a gene trapped mouse that targets one of these isoforms in order to study its biological function. The effect of this gene trap insertion on the splicing of other isoforms has also been studied. RESULTS: In addition to the known 6.1 and 6.2 Kb transcripts of Dlc1, our study revealed the existence of a novel 7.6 Kb transcriptional isoform in the mouse, which corresponds to the human 7.4 Kb (KIAA1723) cDNA transcript. A gene trapped embryonic cell line, with an insertion between Exon 1 and 2 of the 6.1 Kb transcriptional isoform, was used to generate a transgenic mouse. This line showed a significant reduction in the expression of the trapped isoform. However, reduced expression of the other isoforms was not seen. Mice heterozygous for the gene trapped allele were phenotypically normal, but homozygous mutant embryos did not survive beyond 10.5 days post coitum. Dlc1gt/gt embryos showed defects in the brain, heart, and placental blood vessels. Cultured serum-free mouse embryo cells from Dlc1 deficient embryos had elevated RhoA activity and displayed alterations in the organization of actin filaments and focal adhesions. The Dlc1 deficient cells also exhibited increased wound closure in an in vitro scratch assay. CONCLUSIONS: The mouse has three major transcriptional isoforms of the Dlc1 gene that are differentially expressed in various tissues. A mouse with exon 1 of the 6.1 Kb transcript gt resulted in hypomorphic expression of Dlc1 protein and an embryonic lethal phenotype in the homozygous condition, which indicates that this isoform plays a major role in mouse development. The Dlc1 deficient cells showed altered cytoskeleton structure, increased RhoA activity and cellular migration.


Asunto(s)
Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Animales , Northern Blotting , Western Blotting , Línea Celular , Células Cultivadas , Citoesqueleto/metabolismo , Metilación de ADN/genética , Metilación de ADN/fisiología , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Femenino , Genotipo , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Técnicas de Amplificación de Ácido Nucleico , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/fisiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA
11.
Zootaxa ; 5051(1): 7-10, 2021 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-34810909

RESUMEN

John Wells, who died at age 83 on 12 November 2018, was a research scientist, teacher, Professor of Zoology, Dean of Science at Victoria University of Wellington, New Zealand, family man and community leader. He was a man of his time, surviving World War II, progressing to a career that stretched around the world.


Asunto(s)
Zoología , Animales , Historia del Siglo XX , Historia del Siglo XXI , Masculino , Zoología/historia
12.
Elife ; 102021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34723800

RESUMEN

Ataxia Telangiectasia (A-T) and Ataxia with Ocular Apraxia Type 1 (AOA1) are devastating neurological disorders caused by null mutations in the genome stability genes, A-T mutated (ATM) and Aprataxin (APTX), respectively. Our mechanistic understanding and therapeutic repertoire for treating these disorders are severely lacking, in large part due to the failure of prior animal models with similar null mutations to recapitulate the characteristic loss of motor coordination (i.e., ataxia) and associated cerebellar defects. By increasing genotoxic stress through the insertion of null mutations in both the Atm (nonsense) and Aptx (knockout) genes in the same animal, we have generated a novel mouse model that for the first time develops a progressively severe ataxic phenotype associated with atrophy of the cerebellar molecular layer. We find biophysical properties of cerebellar Purkinje neurons (PNs) are significantly perturbed (e.g., reduced membrane capacitance, lower action potential [AP] thresholds, etc.), while properties of synaptic inputs remain largely unchanged. These perturbations significantly alter PN neural activity, including a progressive reduction in spontaneous AP firing frequency that correlates with both cerebellar atrophy and ataxia over the animal's first year of life. Double mutant mice also exhibit a high predisposition to developing cancer (thymomas) and immune abnormalities (impaired early thymocyte development and T-cell maturation), symptoms characteristic of A-T. Finally, by inserting a clinically relevant nonsense-type null mutation in Atm, we demonstrate that Small Molecule Read-Through (SMRT) compounds can restore ATM production, indicating their potential as a future A-T therapeutic.


Asunto(s)
Ataxia Telangiectasia/genética , Atrofia/fisiopatología , Cerebelo/patología , Codón sin Sentido/genética , Células de Purkinje/metabolismo , Animales , Ataxia Telangiectasia/fisiopatología , Atrofia/genética , Modelos Animales de Enfermedad , Femenino , Masculino , Ratones
13.
Cells ; 9(8)2020 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-32784507

RESUMEN

Accurate risk classification of men with localized high-risk prostate cancer directly affects treatment management decisions and patient outcomes. A wide range of risk assessments and classifications are available. However, each one has significant limitations to distinguish between indolent and aggressive prostate cancers. Circulating tumor cells (CTCs) may provide an alternate additional source, beyond tissue biopsies, to enable individual patient-specific clinical assessment, simply because CTCs can reveal both tumor-derived and germline-specific genetic information more precisely than that gained from a single diagnostic biopsy. In this study, we combined a filtration-based CTC isolation technology with prostate cancer CTC immunophenotyping to identify prostate cancer CTCs. Next, we performed 3-D telomere profiling prior to laser microdissection and single-cell whole-exome sequencing (WES) of 21 CTCs and 4 lymphocytes derived from 10 localized high-risk prostate cancer patient samples. Localized high-risk prostate cancer patient CTCs present a high number of telomere signals with lower signal intensities (short telomeres). To capture the genetic diversity/heterogeneity of high-risk prostate cancer CTCs, we carried out whole-exome sequencing. We identified 202,241 single nucleotide variants (SNVs) and 137,407 insertion-deletions (indels), where less than 10% of these genetic variations were within coding regions. The genetic variation (SNVs + indels) and copy number alteration (CNAs) profiles were highly heterogeneous and intra-patient CTC variation was observed. The pathway enrichment analysis showed the presence of genetic variation in nine telomere maintenance pathways (patients 3, 5, 6, and 7), including an important gene for telomere maintenance called telomeric repeat-binding factor 2 (TRF2). Using the PharmGKB database, we identified nine genetic variations associated with response to docetaxel. A total of 48 SNVs can affect drug response for 24 known cancer drugs. Gene Set Enrichment Analysis (GSEA) (patients 1, 3, 6, and 8) identified the presence of CNAs in 11 different pathways, including the DNA damage repair (DDR) pathway. In conclusion, single-cell approaches (WES and 3-D telomere profiling) showed to be useful in unmasking CTC heterogeneity. DDR pathway mutations have been well-established as a target pathway for cancer therapy. However, the frequent CNA amplifications found in localized high-risk patients may play critical roles in the therapeutic resistance in prostate cancer.


Asunto(s)
Variaciones en el Número de Copia de ADN , Células Neoplásicas Circulantes/metabolismo , Polimorfismo de Nucleótido Simple , Neoplasias de la Próstata/genética , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/metabolismo , Genómica , Humanos , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Neoplasias de la Próstata/patología , Análisis de la Célula Individual
14.
Sci Rep ; 9(1): 206, 2019 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-30659253

RESUMEN

We have previously reported the deregulatory impact of ethanol on global DNA methylation of brain-derived neural stem cells (NSC). Here, we conducted a genome-wide RNA-seq analysis in differentiating NSC exposed to different modes of ethanol exposure. RNA-seq results showed distinct gene expression patterns and canonical pathways induced by ethanol exposure and withdrawal. Short-term ethanol exposure caused abnormal up-regulation of synaptic pathways, while continuous ethanol treatment profoundly affected brain cells' morphology. Ethanol withdrawal restored the gene expression profile of differentiating NSC without rescuing impaired expression of epigenetics factors. Ingenuity Pathway Analysis (IPA) analysis predicated that ethanol may impact synaptic functions via GABA receptor signalling pathway and affects neural system and brain morphology. We identified Sptbn2, Dcc, and Scn3a as candidate genes which may link alcohol-induced neuronal morphology to brain structural abnormalities, predicted by IPA analysis. Cross-examination of Scn3a and As3mt in differentiated NSC from two different mouse strains (BL6 and CD1) showed a consistent pattern of induction and reduction, respectively. Collectively, our study identifies genetic networks, which may contribute to alcohol-mediated cellular and brain structural dysmorphology, contributing to our knowledge of alcohol-mediated damage to central nervous system, paving the path for better understanding of FASD pathobiology.


Asunto(s)
Alcoholismo/genética , Etanol/efectos adversos , Efectos Tardíos de la Exposición Prenatal/genética , Alcoholismo/metabolismo , Animales , Encéfalo/metabolismo , Diferenciación Celular/efectos de los fármacos , Depresores del Sistema Nervioso Central/farmacología , Metilación de ADN/efectos de los fármacos , Epigénesis Genética/efectos de los fármacos , Etanol/metabolismo , Etanol/farmacología , Femenino , Perfilación de la Expresión Génica/métodos , Redes Reguladoras de Genes/efectos de los fármacos , Estudio de Asociación del Genoma Completo , Masculino , Ratones , Ratones Endogámicos C57BL/embriología , Ratones Endogámicos/embriología , Canal de Sodio Activado por Voltaje NAV1.3/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Embarazo , Análisis de Secuencia de ARN/métodos , Síndrome de Abstinencia a Sustancias/metabolismo , Transcriptoma/efectos de los fármacos
15.
Carcinogenesis ; 29(4): 722-8, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18258604

RESUMEN

Oxidative stress generated from endogenous and exogenous sources causes oxidative DNA damage. The most frequent mutagenic base lesion 7,8-dihydro-8-oxoguanine and the resulting mismatched adenine are removed by OGG1 and MYH in mammals. Deficiencies in human MYH or mouse MYH and OGG1 result in tumor predisposition but the underlying molecular mechanism is not fully understood. To facilitate the study of the roles of MYH and OGG1 in the protection against oxidative stress, we generated mouse embryonic fibroblast cell lines deficient in these genes. Myh and Ogg1 double knockout cells were more sensitive than wild type to oxidants (hydrogen peroxide and t-butyl hydroperoxide), but not to cis-platinum or gamma-irradiations. The low dosage oxidative stress resulted in more reduction of S phase and increase of G(2)/M phase in Myh(-/-)Ogg1(-/-) cells than in wild-type cells, but a similar level of cell death in both cells. The oxidants also induced more multinucleated cells in Myh(-/-)Ogg1(-/-) cells than in wild-type, accompanied by centrosome amplification and multipolar spindle formation. Thus, under oxidative stress, Myh and Ogg1 are likely required for normal cell-cycle progression and nuclear division, suggesting multiple roles of Myh and Ogg1 in the maintenance of genome stability and tumor prevention.


Asunto(s)
División Celular/efectos de los fármacos , Daño del ADN , ADN Glicosilasas/deficiencia , Fase G2/efectos de los fármacos , Oxidantes/toxicidad , Estrés Oxidativo , Animales , Cruzamientos Genéticos , Femenino , Humanos , Peróxido de Hidrógeno/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , terc-Butilhidroperóxido/farmacología
16.
Curr Biol ; 15(6): 587-93, 2005 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-15797031

RESUMEN

Neuronal dendrites, together with dendritic spines, exhibit enormously diverse structure. Selective targeting and local translation of mRNAs in dendritic spines have been implicated in synapse remodeling or synaptic plasticity. The mechanism of mRNA transport to the postsynaptic site is a fundamental question in local dendritic translation. TLS (translocated in liposarcoma), previously identified as a component of hnRNP complexes, unexpectedly showed somatodendritic localization in mature hippocampal pyramidal neurons. In the present study, TLS was translocated to dendrites and was recruited to dendrites not only via microtubules but also via actin filaments. In mature hippocampal pyramidal neurons, TLS accumulated in the spines at excitatory postsynapses upon mGluR5 activation, which was accompanied by an increased RNA content in dendrites. Consistent with the in vitro studies, TLS-null hippocampal pyramidal neurons exhibited abnormal spine morphology and lower spine density. Our results indicate that TLS participates in mRNA sorting to the dendritic spines induced by mGluR5 activation and regulates spine morphology to stabilize the synaptic structure.


Asunto(s)
Espinas Dendríticas/metabolismo , Hipocampo/metabolismo , Neuronas/citología , ARN Mensajero/metabolismo , Proteína FUS de Unión a ARN/metabolismo , Receptores de Ácido Kaínico/metabolismo , Actinas/metabolismo , Animales , Células Cultivadas , Recuperación de Fluorescencia tras Fotoblanqueo , Proteínas Fluorescentes Verdes , Inmunohistoquímica , Ratones , Microscopía Confocal , Microtúbulos/metabolismo , Neuronas/metabolismo , Transporte de Proteínas/fisiología
17.
Nucleic Acids Res ; 34(Database issue): D642-8, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16381950

RESUMEN

Gene trapping is a method of generating murine embryonic stem (ES) cell lines containing insertional mutations in known and novel genes. A number of international groups have used this approach to create sizeable public cell line repositories available to the scientific community for the generation of mutant mouse strains. The major gene trapping groups worldwide have recently joined together to centralize access to all publicly available gene trap lines by developing a user-oriented Website for the International Gene Trap Consortium (IGTC). This collaboration provides an impressive public informatics resource comprising approximately 45 000 well-characterized ES cell lines which currently represent approximately 40% of known mouse genes, all freely available for the creation of knockout mice on a non-collaborative basis. To standardize annotation and provide high confidence data for gene trap lines, a rigorous identification and annotation pipeline has been developed combining genomic localization and transcript alignment of gene trap sequence tags to identify trapped loci. This information is stored in a new bioinformatics database accessible through the IGTC Website interface. The IGTC Website (www.genetrap.org) allows users to browse and search the database for trapped genes, BLAST sequences against gene trap sequence tags, and view trapped genes within biological pathways. In addition, IGTC data have been integrated into major genome browsers and bioinformatics sites to provide users with outside portals for viewing this data. The development of the IGTC Website marks a major advance by providing the research community with the data and tools necessary to effectively use public gene trap resources for the large-scale characterization of mammalian gene function.


Asunto(s)
Línea Celular , Bases de Datos de Ácidos Nucleicos , Ratones/genética , Mutagénesis Insercional , Animales , Mapeo Cromosómico , Embrión de Mamíferos/citología , Cooperación Internacional , Internet , Ratones/embriología , Ratones Noqueados , Mutagénesis Insercional/métodos , ARN Mensajero/análisis , Integración de Sistemas , Interfaz Usuario-Computador
18.
Neuron ; 100(4): 816-830.e7, 2018 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-30344044

RESUMEN

Through the generation of humanized FUS mice expressing full-length human FUS, we identify that when expressed at near endogenous murine FUS levels, both wild-type and ALS-causing and frontotemporal dementia (FTD)-causing mutations complement the essential function(s) of murine FUS. Replacement of murine FUS with mutant, but not wild-type, human FUS causes stress-mediated induction of chaperones, decreased expression of ion channels and transporters essential for synaptic function, and reduced synaptic activity without loss of nuclear FUS or its cytoplasmic aggregation. Most strikingly, accumulation of mutant human FUS is shown to activate an integrated stress response and to inhibit local, intra-axonal protein synthesis in hippocampal neurons and sciatic nerves. Collectively, our evidence demonstrates that human ALS/FTD-linked mutations in FUS induce a gain of toxicity that includes stress-mediated suppression in intra-axonal translation, synaptic dysfunction, and progressive age-dependent motor and cognitive disease without cytoplasmic aggregation, altered nuclear localization, or aberrant splicing of FUS-bound pre-mRNAs. VIDEO ABSTRACT.


Asunto(s)
Esclerosis Amiotrófica Lateral/genética , Axones/fisiología , Demencia Frontotemporal/genética , Mutación con Pérdida de Función/genética , Biosíntesis de Proteínas/fisiología , Proteína FUS de Unión a ARN/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Axones/patología , Células Cultivadas , Femenino , Demencia Frontotemporal/metabolismo , Demencia Frontotemporal/patología , Hipocampo/metabolismo , Hipocampo/patología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo , Proteína FUS de Unión a ARN/biosíntesis
19.
Mol Cell Biol ; 23(15): 5301-7, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12861016

RESUMEN

The high-mobility-group (HMG) SSRP1 protein is a member of a conserved chromatin-remodeling complex (FACT/DUF/CP) implicated in DNA replication, basal and regulated transcription, and DNA repair. To assist in the functional analysis of SSRP1, the Ssrp1 gene was targeted in murine embryonic stem cells, and the mutation was introduced into the germ line. Embryos homozygous for the targeted allele die soon after implantation, and preimplantation blastocysts are defective for cell outgrowth and/or survival in vitro. The Ssrp1 mutation was also crossed into a p53 null background without affecting growth and/or survival defects caused by loss of Ssrp1 function. Thus, Ssrp1 appears to encode nonredundant and p53-independent functions that are essential for cell viability.


Asunto(s)
Proteínas de Unión al ADN/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Alelos , Animales , Apoptosis , Southern Blotting , Western Blotting , División Celular , Supervivencia Celular , Proteínas de Unión al ADN/química , Genes p53 , Vectores Genéticos , Genotipo , Proteínas del Grupo de Alta Movilidad/química , Homocigoto , Etiquetado Corte-Fin in Situ , Ratones , Modelos Genéticos , Mutación , Recombinación Genética , Células Madre/metabolismo , Factores de Tiempo
20.
Sci Rep ; 7(1): 6382, 2017 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-28743913

RESUMEN

miR-200b plays a role in epithelial-to-mesenchymal transition (EMT) in cancer. We recently reported abnormal expression of miR-200b in the context of human pulmonary hypoplasia in congenital diaphragmatic hernia (CDH). Smaller lung size, a lower number of airway generations, and a thicker mesenchyme characterize pulmonary hypoplasia in CDH. The aim of this study was to define the role of miR-200b during lung development. Here we show that miR-200b-/- mice have abnormal lung function due to dysfunctional surfactant, increased fibroblast-like cells and thicker mesenchyme in between the alveolar walls. We profiled the lung transcriptome in miR-200b-/- mice, and, using Gene Ontology analysis, we determined that the most affected biological processes include cell cycle, apoptosis and protein transport. Our results demonstrate that miR-200b regulates distal airway development through maintaining an epithelial cell phenotype. The lung abnormalities observed in miR-200b-/- mice recapitulate lung hypoplasia in CDH.


Asunto(s)
Células Epiteliales/citología , Pulmón/crecimiento & desarrollo , MicroARNs/genética , Regulación hacia Arriba , Animales , Células Epiteliales/patología , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Ontología de Genes , Redes Reguladoras de Genes , Hernias Diafragmáticas Congénitas/genética , Hernias Diafragmáticas Congénitas/fisiopatología , Humanos , Pulmón/citología , Pulmón/fisiopatología , Ratones , Pruebas de Función Respiratoria , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA