Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Biol Evol ; 40(9)2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37739926

RESUMEN

Fins are major functional appendages of fish that have been repeatedly modified in different lineages. To search for genomic changes underlying natural fin diversity, we compared the genomes of 36 percomorph fish species that span over 100 million years of evolution and either have complete or reduced pelvic and caudal fins. We identify 1,614 genomic regions that are well-conserved in fin-complete species but missing from multiple fin-reduced lineages. Recurrent deletions of conserved sequences in wild fin-reduced species are enriched for functions related to appendage development, suggesting that convergent fin reduction at the organismal level is associated with repeated genomic deletions near fin-appendage development genes. We used sequencing and functional enhancer assays to confirm that PelA, a Pitx1 enhancer previously linked to recurrent pelvic loss in sticklebacks, has also been independently deleted and may have contributed to the fin morphology in distantly related pelvic-reduced species. We also identify a novel enhancer that is conserved in the majority of percomorphs, drives caudal fin expression in transgenic stickleback, is missing in tetraodontiform, syngnathid, and synbranchid species with caudal fin reduction, and alters caudal fin development when targeted by genome editing. Our study illustrates a broadly applicable strategy for mapping phenotypes to genotypes across a tree of vertebrate species and highlights notable new examples of regulatory genomic hotspots that have been used to evolve recurrent phenotypes across 100 million years of fish evolution.


Asunto(s)
Peces , Smegmamorpha , Animales , Peces/genética , Genómica , Genotipo , Smegmamorpha/genética , Aletas de Animales
2.
BMC Genomics ; 24(1): 97, 2023 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-36864393

RESUMEN

BACKGROUND: 'Long read' sequencing methods have been used to identify previously uncharacterized structural variants that cause human genetic diseases. Therefore, we investigated whether long read sequencing could facilitate genetic analysis of murine models for human diseases. RESULTS: The genomes of six inbred strains (BTBR T + Itpr3tf/J, 129Sv1/J, C57BL/6/J, Balb/c/J, A/J, SJL/J) were analyzed using long read sequencing. Our results revealed that (i) Structural variants are very abundant within the genome of inbred strains (4.8 per gene) and (ii) that we cannot accurately infer whether structural variants are present using conventional short read genomic sequence data, even when nearby SNP alleles are known. The advantage of having a more complete map was demonstrated by analyzing the genomic sequence of BTBR mice. Based upon this analysis, knockin mice were generated and used to characterize a BTBR-unique 8-bp deletion within Draxin that contributes to the BTBR neuroanatomic abnormalities, which resemble human autism spectrum disorder. CONCLUSION: A more complete map of the pattern of genetic variation among inbred strains, which is produced by long read genomic sequencing of the genomes of additional inbred strains, could facilitate genetic discovery when murine models of human diseases are analyzed.


Asunto(s)
Trastorno del Espectro Autista , Humanos , Ratones , Animales , Ratones Endogámicos C57BL , Ratones Endogámicos , Mapeo Cromosómico , Alelos , Péptidos y Proteínas de Señalización Intercelular
3.
Development ; 147(24)2020 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-33144399

RESUMEN

Sense organs acquire their distinctive shapes concomitantly with the differentiation of sensory cells and neurons necessary for their function. Although our understanding of the mechanisms controlling morphogenesis and neurogenesis in these structures has grown, how these processes are coordinated remains largely unexplored. Neurogenesis in the zebrafish olfactory epithelium requires the bHLH proneural transcription factor Neurogenin 1 (Neurog1). To address whether Neurog1 also controls morphogenesis, we analysed the migratory behaviour of early olfactory neural progenitors in neurog1 mutant embryos. Our results indicate that the oriented movements of these progenitors are disrupted in this context. Morphogenesis is similarly affected by mutations in the chemokine receptor gene, cxcr4b, suggesting it is a potential Neurog1 target gene. We find that Neurog1 directly regulates cxcr4b through an E-box cluster located just upstream of the cxcr4b transcription start site. Our results suggest that proneural transcription factors, such as Neurog1, directly couple distinct aspects of nervous system development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Morfogénesis/genética , Proteínas del Tejido Nervioso/genética , Neurogénesis/genética , Mucosa Olfatoria/crecimiento & desarrollo , Receptores CXCR4/genética , Proteínas de Pez Cebra/genética , Animales , Elementos E-Box/genética , Embrión no Mamífero , Desarrollo Embrionario/genética , Regulación del Desarrollo de la Expresión Génica/genética , Mutación/genética , Neuronas/metabolismo , Sitio de Iniciación de la Transcripción , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
4.
PLoS Comput Biol ; 18(8): e1010378, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-36040971

RESUMEN

We present WhichTF, a computational method to identify functionally important transcription factors (TFs) from chromatin accessibility measurements. To rank TFs, WhichTF applies an ontology-guided functional approach to compute novel enrichment by integrating accessibility measurements, high-confidence pre-computed conservation-aware TF binding sites, and putative gene-regulatory models. Comparison with prior sheer abundance-based methods reveals the unique ability of WhichTF to identify context-specific TFs with functional relevance, including NF-κB family members in lymphocytes and GATA factors in cardiac cells. To distinguish the transcriptional regulatory landscape in closely related samples, we apply differential analysis and demonstrate its utility in lymphocyte, mesoderm developmental, and disease cells. We find suggestive, under-characterized TFs, such as RUNX3 in mesoderm development and GLI1 in systemic lupus erythematosus. We also find TFs known for stress response, suggesting routine experimental caveats that warrant careful consideration. WhichTF yields biological insight into known and novel molecular mechanisms of TF-mediated transcriptional regulation in diverse contexts, including human and mouse cell types, cell fate trajectories, and disease-associated cells.


Asunto(s)
Cromatina , Factores de Transcripción , Animales , Sitios de Unión , Cromatina/genética , Regulación de la Expresión Génica , Humanos , Ratones , Unión Proteica , Factores de Transcripción/metabolismo
5.
Proc Natl Acad Sci U S A ; 117(40): 25074-25084, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32948690

RESUMEN

We are only just beginning to catalog the vast diversity of cell types in the cerebral cortex. Such categorization is a first step toward understanding how diversification relates to function. All cortical projection neurons arise from a uniform pool of progenitor cells that lines the ventricles of the forebrain. It is still unclear how these progenitor cells generate the more than 50 unique types of mature cortical projection neurons defined by their distinct gene-expression profiles. Moreover, exactly how and when neurons diversify their function during development is unknown. Here we relate gene expression and chromatin accessibility of two subclasses of projection neurons with divergent morphological and functional features as they develop in the mouse brain between embryonic day 13 and postnatal day 5 in order to identify transcriptional networks that diversify neuron cell fate. We compare these gene-expression profiles with published profiles of single cells isolated from similar populations and establish that layer-defined cell classes encompass cell subtypes and developmental trajectories identified using single-cell sequencing. Given the depth of our sequencing, we identify groups of transcription factors with particularly dense subclass-specific regulation and subclass-enriched transcription factor binding motifs. We also describe transcription factor-adjacent long noncoding RNAs that define each subclass and validate the function of Myt1l in balancing the ratio of the two subclasses in vitro. Our multidimensional approach supports an evolving model of progressive restriction of cell fate competence through inherited transcriptional identities.


Asunto(s)
Proteínas del Tejido Nervioso/genética , Neuronas/metabolismo , Análisis de la Célula Individual , Factores de Transcripción/genética , Animales , Diferenciación Celular/genética , Corteza Cerebral/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Ratones , RNA-Seq/métodos
6.
Genet Med ; 24(10): 2091-2102, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35976265

RESUMEN

PURPOSE: Cohort building is a powerful foundation for improving clinical care, performing biomedical research, recruiting for clinical trials, and many other applications. We set out to build a cohort of all monogenic patients with a definitive causal gene diagnosis in a 3-million patient hospital system. METHODS: We define a subset (4461) of OMIM diseases that have at least 1 known monogenic causal gene. We then introduce MonoMiner, a natural language processing framework to identify molecularly confirmed monogenic patients from free-text clinical notes. RESULTS: We show that ICD-10-CM codes cover only a fraction of monogenic diseases and that even where available, ICD-10-CM code‒based patient retrieval offers 0.14 precision. Searching by causal gene symbol offers great recall but has an even worse 0.07 precision. MonoMiner achieves 6 to 11 times higher precision (0.80), with 0.87 precision on disease diagnosis alone, tagging 4259 patients with 560 monogenic diseases and 534 causal genes, at 0.48 recall. CONCLUSION: MonoMiner enables the discovery of a large, high-precision cohort of patients with monogenic diseases with an established molecular diagnosis, empowering numerous downstream uses. Because it relies solely on clinical notes, MonoMiner is highly portable, and its approach is adaptable to other domains and languages.


Asunto(s)
Registros Electrónicos de Salud , Procesamiento de Lenguaje Natural , Estudios de Cohortes , Humanos
7.
Nucleic Acids Res ; 48(16): e91, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32614390

RESUMEN

Gene losses provide an insightful route for studying the morphological and physiological adaptations of species, but their discovery is challenging. Existing genome annotation tools focus on annotating intact genes and do not attempt to distinguish nonfunctional genes from genes missing annotation due to sequencing and assembly artifacts. Previous attempts to annotate gene losses have required significant manual curation, which hampers their scalability for the ever-increasing deluge of newly sequenced genomes. Using extreme sequence erosion (amino acid deletions and substitutions) and sister species support as an unambiguous signature of loss, we developed an automated approach for detecting high-confidence gene loss events across a species tree. Our approach relies solely on gene annotation in a single reference genome, raw assemblies for the remaining species to analyze, and the associated phylogenetic tree for all organisms involved. Using human as reference, we discovered over 400 unique human ortholog erosion events across 58 mammals. This includes dozens of clade-specific losses of genes that result in early mouse lethality or are associated with severe human congenital diseases. Our discoveries yield intriguing potential for translational medical genetics and evolutionary biology, and our approach is readily applicable to large-scale genome sequencing efforts across the tree of life.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Genómica/métodos , Filogenia , Algoritmos , Animales , Automatización , Mapeo Cromosómico/métodos , Genes Letales , Humanos , Mamíferos/genética , Ratones , Anotación de Secuencia Molecular
8.
Proc Natl Acad Sci U S A ; 116(42): 21094-21103, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31570615

RESUMEN

Distantly related species entering similar biological niches often adapt by evolving similar morphological and physiological characters. How much genomic molecular convergence (particularly of highly constrained coding sequence) contributes to convergent phenotypic evolution, such as echolocation in bats and whales, is a long-standing fundamental question. Like others, we find that convergent amino acid substitutions are not more abundant in echolocating mammals compared to their outgroups. However, we also ask a more informative question about the genomic distribution of convergent substitutions by devising a test to determine which, if any, of more than 4,000 tissue-affecting gene sets is most statistically enriched with convergent substitutions. We find that the gene set most overrepresented (q-value = 2.2e-3) with convergent substitutions in echolocators, affecting 18 genes, regulates development of the cochlear ganglion, a structure with empirically supported relevance to echolocation. Conversely, when comparing to nonecholocating outgroups, no significant gene set enrichment exists. For aquatic and high-altitude mammals, our analysis highlights 15 and 16 genes from the gene sets most affected by molecular convergence which regulate skin and lung physiology, respectively. Importantly, our test requires that the most convergence-enriched set cannot also be enriched for divergent substitutions, such as in the pattern produced by inactivated vision genes in subterranean mammals. Showing a clear role for adaptive protein-coding molecular convergence, we discover nearly 2,600 convergent positions, highlight 77 of them in 3 organs, and provide code to investigate other clades across the tree of life.


Asunto(s)
Quirópteros/genética , Quirópteros/fisiología , Ecolocación/fisiología , Proteínas/genética , Ballenas/genética , Ballenas/fisiología , Adaptación Fisiológica/genética , Adaptación Fisiológica/fisiología , Sustitución de Aminoácidos/genética , Animales , Evolución Molecular , Genoma/genética , Genómica/métodos , Audición/genética , Audición/fisiología , Filogenia , Selección Genética/genética
9.
Genet Med ; 23(10): 1984-1992, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34230641

RESUMEN

PURPOSE: Roughly 70% of suspected Mendelian disease patients remain undiagnosed after genome sequencing, partly because knowledge about pathogenic genes is incomplete and constantly growing. Generating a novel pathogenic gene hypothesis from patient data can be time-consuming especially where cohort-based analysis is not available. METHODS: Each patient genome contains dozens to hundreds of candidate variants. Many sources of indirect evidence about each candidate may be considered. We introduce InpherNet, a network-based machine learning approach leveraging Monarch Initiative data to accelerate this process. RESULTS: InpherNet ranks candidate genes based on orthologs, paralogs, functional pathway members, and colocalized interaction partner gene neighbors. It can propose novel pathogenic genes and reveal known pathogenic genes whose diagnosed patient-based annotation is missing or partial. InpherNet is applied to patient cases where the causative gene is incorrectly ranked low by clinical gene-ranking methods that use only patient-derived evidence. InpherNet correctly ranks the causative gene top 1 or top 1-5 in roughly twice as many cases as seven comparable tools, including in cases where no clinical evidence for the diagnostic gene is in our knowledgebase. CONCLUSION: InpherNet improves the state of the art in considering candidate gene neighbors to accelerate monogenic diagnosis.


Asunto(s)
Enfermedades Genéticas Congénitas/diagnóstico , Bases del Conocimiento , Aprendizaje Automático , Estudios de Cohortes , Humanos
10.
RNA ; 24(12): 1647-1658, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30224349

RESUMEN

Experimental detection of RNA splicing branchpoints is difficult. To date, high-confidence experimental annotations exist for 18% of 3' splice sites in the human genome. We develop a deep-learning-based branchpoint predictor, LaBranchoR, which predicts a correct branchpoint for at least 75% of 3' splice sites genome-wide. Detailed analysis of cases in which our predicted branchpoint deviates from experimental data suggests a correct branchpoint is predicted in over 90% of cases. We use our predicted branchpoints to identify a novel sequence element upstream of branchpoints consistent with extended U2 snRNA base-pairing, show an association between weak branchpoints and alternative splicing, and explore the effects of genetic variants on branchpoints. We provide genome-wide branchpoint annotations and in silico mutagenesis scores at http://bejerano.stanford.edu/labranchor.


Asunto(s)
Empalme Alternativo/genética , Genoma Humano/genética , Empalme del ARN/genética , ARN Nuclear Pequeño/genética , Simulación por Computador , Aprendizaje Profundo , Exones/genética , Humanos , Intrones/genética , Anotación de Secuencia Molecular , Mutagénesis/genética , Sitios de Empalme de ARN/genética
11.
Genet Med ; 22(2): 362-370, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31467448

RESUMEN

PURPOSE: Both monogenic pathogenic variant cataloging and clinical patient diagnosis start with variant-level evidence retrieval followed by expert evidence integration in search of diagnostic variants and genes. Here, we try to accelerate pathogenic variant evidence retrieval by an automatic approach. METHODS: Automatic VAriant evidence DAtabase (AVADA) is a novel machine learning tool that uses natural language processing to automatically identify pathogenic genetic variant evidence in full-text primary literature about monogenic disease and convert it to genomic coordinates. RESULTS: AVADA automatically retrieved almost 60% of likely disease-causing variants deposited in the Human Gene Mutation Database (HGMD), a 4.4-fold improvement over the current best open source automated variant extractor. AVADA contains over 60,000 likely disease-causing variants that are in HGMD but not in ClinVar. AVADA also highlights the challenges of automated variant mapping and pathogenicity curation. However, when combined with manual validation, on 245 diagnosed patients, AVADA provides valuable evidence for an additional 18 diagnostic variants, on top of ClinVar's 21, versus only 2 using the best current automated approach. CONCLUSION: AVADA advances automated retrieval of pathogenic monogenic variant evidence from full-text literature. Far from perfect, but much faster than PubMed/Google Scholar search, careful curation of AVADA-retrieved evidence can aid both database curation and patient diagnosis.


Asunto(s)
Procesamiento Automatizado de Datos/métodos , Genómica/métodos , Almacenamiento y Recuperación de la Información/métodos , Manejo de Datos/métodos , Bases de Datos Factuales , Bases de Datos Genéticas , Humanos , Procesamiento de Lenguaje Natural , PubMed , Publicaciones
12.
Nucleic Acids Res ; 46(18): 9299-9308, 2018 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-30137416

RESUMEN

Genetic variation in cis-regulatory elements is thought to be a major driving force in morphological and physiological changes. However, identifying transcription factor binding events that code for complex traits remains a challenge, motivating novel means of detecting putatively important binding events. Using a curated set of 1154 high-quality transcription factor motifs, we demonstrate that independently eroded binding sites are enriched for independently lost traits in three distinct pairs of placental mammals. We show that these independently eroded events pinpoint the loss of hindlimbs in dolphin and manatee, degradation of vision in naked mole-rat and star-nosed mole, and the loss of external testes in white rhinoceros and Weddell seal. We additionally show that our method may also be utilized with more than two species. Our study exhibits a novel methodology to detect cis-regulatory mutations which help explain a portion of the molecular mechanism underlying complex trait formation and loss.


Asunto(s)
Evolución Molecular , Motivos de Nucleótidos/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/genética , Visión Ocular/genética , Animales , Sitios de Unión/genética , Delfines/genética , Delfines/fisiología , Miembro Posterior/fisiología , Masculino , Mamíferos/genética , Mamíferos/fisiología , Ratas Topo/genética , Ratas Topo/fisiología , Unión Proteica/genética , Testículo/fisiología , Trichechus/genética , Trichechus/fisiología , Visión Ocular/fisiología
13.
Nucleic Acids Res ; 46(7): 3517-3531, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29518216

RESUMEN

Thousands of human disease-associated single nucleotide polymorphisms (SNPs) lie in the non-coding genome, but only a handful have been demonstrated to affect gene expression and human biology. We computationally identified risk-associated SNPs in deeply conserved non-exonic elements (CNEs) potentially contributing to 45 human diseases. We further demonstrated that human CNE1/rs17421627 associated with retinal vasculature defects showed transcriptional activity in the zebrafish retina, while introducing the risk-associated allele completely abolished CNE1 enhancer activity. Furthermore, deletion of CNE1 led to retinal vasculature defects and to a specific downregulation of microRNA-9, rather than MEF2C as predicted by the original genome-wide association studies. Consistent with these results, miR-9 depletion affects retinal vasculature formation, demonstrating MIR-9-2 as a critical gene underpinning the associated trait. Importantly, we validated that other CNEs act as transcriptional enhancers that can be disrupted by conserved non-coding SNPs. This study uncovers disease-associated non-coding mutations that are deeply conserved, providing a path for in vivo testing to reveal their cis-regulated genes and biological roles.


Asunto(s)
Elementos de Facilitación Genéticos/genética , MicroARNs/genética , Vasculitis Retiniana/genética , Alelos , Animales , Secuencia Conservada/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Estudio de Asociación del Genoma Completo , Humanos , Factores de Transcripción MEF2/genética , Mutación , Polimorfismo de Nucleótido Simple/genética , Retina/metabolismo , Retina/patología , Vasculitis Retiniana/patología , Pez Cebra/genética
14.
Genome Res ; 26(8): 1013-22, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27325115

RESUMEN

Exome sequencing studies have identified multiple genes harboring de novo loss-of-function (LoF) variants in individuals with autism spectrum disorders (ASD), including TBR1, a master regulator of cortical development. We performed ChIP-seq for TBR1 during mouse cortical neurogenesis and show that TBR1-bound regions are enriched adjacent to ASD genes. ASD genes were also enriched among genes that are differentially expressed in Tbr1 knockouts, which together with the ChIP-seq data, suggests direct transcriptional regulation. Of the nine ASD genes examined, seven were misexpressed in the cortices of Tbr1 knockout mice, including six with increased expression in the deep cortical layers. ASD genes with adjacent cortical TBR1 ChIP-seq peaks also showed unusually low levels of LoF mutations in a reference human population and among Icelanders. We then leveraged TBR1 binding to identify an appealing subset of candidate ASD genes. Our findings highlight a TBR1-regulated network of ASD genes in the developing neocortex that are relatively intolerant to LoF mutations, indicating that these genes may play critical roles in normal cortical development.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas de Unión al ADN/genética , Neocórtex/fisiopatología , Neurogénesis/genética , Animales , Trastorno del Espectro Autista/fisiopatología , Modelos Animales de Enfermedad , Exoma/genética , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Humanos , Ratones , Mutación , Neocórtex/crecimiento & desarrollo , Neuronas/metabolismo , Neuronas/patología , Factores de Riesgo , Proteínas de Dominio T Box
15.
Genet Med ; 21(2): 464-470, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29997393

RESUMEN

PURPOSE: Exome sequencing and diagnosis is beginning to spread across the medical establishment. The most time-consuming part of genome-based diagnosis is the manual step of matching the potentially long list of patient candidate genes to patient phenotypes to identify the causative disease. METHODS: We introduce Phrank (for phenotype ranking), an information theory-inspired method that utilizes a Bayesian network to prioritize candidate diseases or genes, as a stand-alone module that can be run with any underlying knowledgebase and any variant filtering scheme. RESULTS: Phrank outperforms existing methods at ranking the causative disease or gene when applied to 169 real patient exomes with Mendelian diagnoses. Phrank's greatest improvement is in disease space, where across all 169 patients it ranks only 3 diseases on average ahead of the true diagnosis, whereas Phenomizer ranks 32 diseases ahead of the causal one. CONCLUSIONS: Using Phrank to rank all patient candidate genes or diseases, as they start working through a new case, will save the busy clinician much time in deriving a genetic diagnosis.


Asunto(s)
Diagnóstico por Computador , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas , Fenotipo , Programas Informáticos , Benchmarking , Biología Computacional/métodos , Exoma , Humanos , Bases del Conocimiento , Patología Molecular/métodos
16.
Genet Med ; 21(7): 1585-1593, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30514889

RESUMEN

PURPOSE: Diagnosing monogenic diseases facilitates optimal care, but can involve the manual evaluation of hundreds of genetic variants per case. Computational tools like Phrank expedite this process by ranking all candidate genes by their ability to explain the patient's phenotypes. To use these tools, busy clinicians must manually encode patient phenotypes from lengthy clinical notes. With 100 million human genomes estimated to be sequenced by 2025, a fast alternative to manual phenotype extraction from clinical notes will become necessary. METHODS: We introduce ClinPhen, a fast, high-accuracy tool that automatically converts clinical notes into a prioritized list of patient phenotypes using Human Phenotype Ontology (HPO) terms. RESULTS: ClinPhen shows superior accuracy and 20× speedup over existing phenotype extractors, and its novel phenotype prioritization scheme improves the performance of gene-ranking tools. CONCLUSION: While a dedicated clinician can process 200 patient records in a 40-hour workweek, ClinPhen does the same in 10 minutes. Compared with manual phenotype extraction, ClinPhen saves an additional 3-5 hours per Mendelian disease diagnosis. Providers can now add ClinPhen's output to each summary note attached to a filled testing laboratory request form. ClinPhen makes a substantial contribution to improvements in efficiency critically needed to meet the surging demand for clinical diagnostic sequencing.


Asunto(s)
Biología Computacional , Enfermedades Genéticas Congénitas/diagnóstico , Registros Médicos , Algoritmos , Humanos , Procesamiento de Lenguaje Natural , Fenotipo
17.
Nat Rev Genet ; 14(4): 288-95, 2013 04.
Artículo en Inglés | MEDLINE | ID: mdl-23503198

RESUMEN

It is estimated that the human genome contains hundreds of thousands of enhancers, so understanding these gene-regulatory elements is a crucial goal. Several fundamental questions need to be addressed about enhancers, such as how do we identify them all, how do they work, and how do they contribute to disease and evolution? Five prominent researchers in this field look at how much we know already and what needs to be done to answer these questions.


Asunto(s)
Elementos de Facilitación Genéticos/genética , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad/genética , Regiones Promotoras Genéticas/genética , Evolución Molecular , Humanos , Mutación
18.
Am J Med Genet A ; 176(4): 1030-1036, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29575631

RESUMEN

Robinow syndrome (RS) is a well-recognized Mendelian disorder known to demonstrate both autosomal dominant and autosomal recessive inheritance. Typical manifestations include short stature, characteristic facies, and skeletal anomalies. Recessive inheritance has been associated with mutations in ROR2 while dominant inheritance has been observed for mutations in WNT5A, DVL1, and DVL3. Through trio whole genome sequencing, we identified a homozygous frameshifting single nucleotide deletion in WNT5A in a previously reported, deceased infant with a unique constellation of features comprising a 46,XY disorder of sex development with multiple congenital malformations including congenital diaphragmatic hernia, ambiguous genitalia, dysmorphic facies, shortened long bones, adactyly, and ventricular septal defect. The parents, who are both heterozygous for the deletion, appear clinically unaffected. In conjunction with published observations of Wnt5a double knockout mice, we provide evidence for the possibility of autosomal recessive inheritance in association with WNT5A loss-of-function mutations in RS.


Asunto(s)
Alelos , Anomalías Craneofaciales/diagnóstico , Anomalías Craneofaciales/genética , Enanismo/diagnóstico , Enanismo/genética , Deformidades Congénitas de las Extremidades/diagnóstico , Deformidades Congénitas de las Extremidades/genética , Mutación con Pérdida de Función , Fenotipo , Anomalías Urogenitales/diagnóstico , Anomalías Urogenitales/genética , Proteína Wnt-5a/genética , Animales , Modelos Animales de Enfermedad , Femenino , Mutación del Sistema de Lectura , Frecuencia de los Genes , Estudios de Asociación Genética , Homocigoto , Humanos , Lactante , Ratones , Ratones Noqueados , Mutación Puntual , Índice de Severidad de la Enfermedad , Evaluación de Síntomas , Ultrasonografía , Secuenciación Completa del Genoma
19.
Brain ; 140(10): 2610-2622, 2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28969385

RESUMEN

Mutations of genes within the phosphatidylinositol-3-kinase (PI3K)-AKT-MTOR pathway are well known causes of brain overgrowth (megalencephaly) as well as segmental cortical dysplasia (such as hemimegalencephaly, focal cortical dysplasia and polymicrogyria). Mutations of the AKT3 gene have been reported in a few individuals with brain malformations, to date. Therefore, our understanding regarding the clinical and molecular spectrum associated with mutations of this critical gene is limited, with no clear genotype-phenotype correlations. We sought to further delineate this spectrum, study levels of mosaicism and identify genotype-phenotype correlations of AKT3-related disorders. We performed targeted sequencing of AKT3 on individuals with these phenotypes by molecular inversion probes and/or Sanger sequencing to determine the type and level of mosaicism of mutations. We analysed all clinical and brain imaging data of mutation-positive individuals including neuropathological analysis in one instance. We performed ex vivo kinase assays on AKT3 engineered with the patient mutations and examined the phospholipid binding profile of pleckstrin homology domain localizing mutations. We identified 14 new individuals with AKT3 mutations with several phenotypes dependent on the type of mutation and level of mosaicism. Our comprehensive clinical characterization, and review of all previously published patients, broadly segregates individuals with AKT3 mutations into two groups: patients with highly asymmetric cortical dysplasia caused by the common p.E17K mutation, and patients with constitutional AKT3 mutations exhibiting more variable phenotypes including bilateral cortical malformations, polymicrogyria, periventricular nodular heterotopia and diffuse megalencephaly without cortical dysplasia. All mutations increased kinase activity, and pleckstrin homology domain mutants exhibited enhanced phospholipid binding. Overall, our study shows that activating mutations of the critical AKT3 gene are associated with a wide spectrum of brain involvement ranging from focal or segmental brain malformations (such as hemimegalencephaly and polymicrogyria) predominantly due to mosaic AKT3 mutations, to diffuse bilateral cortical malformations, megalencephaly and heterotopia due to constitutional AKT3 mutations. We also provide the first detailed neuropathological examination of a child with extreme megalencephaly due to a constitutional AKT3 mutation. This child has one of the largest documented paediatric brain sizes, to our knowledge. Finally, our data show that constitutional AKT3 mutations are associated with megalencephaly, with or without autism, similar to PTEN-related disorders. Recognition of this broad clinical and molecular spectrum of AKT3 mutations is important for providing early diagnosis and appropriate management of affected individuals, and will facilitate targeted design of future human clinical trials using PI3K-AKT pathway inhibitors.


Asunto(s)
Discapacidades del Desarrollo/genética , Megalencefalia/genética , Mutación/genética , Proteínas Proto-Oncogénicas c-akt/genética , Encéfalo/diagnóstico por imagen , Niño , Discapacidades del Desarrollo/diagnóstico por imagen , Discapacidades del Desarrollo/patología , Femenino , Estudios de Asociación Genética , Células HEK293 , Humanos , Inmunoprecipitación , Imagen por Resonancia Magnética , Masculino , Megalencefalia/diagnóstico por imagen , Megalencefalia/patología , Mutagénesis Sitio-Dirigida/métodos , Fosfatidilinositoles/metabolismo , Transfección
20.
Proc Natl Acad Sci U S A ; 112(26): 8036-40, 2015 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-26080416

RESUMEN

Viral outbreaks in dolphins and other Delphinoidea family members warrant investigation into the integrity of the cetacean immune system. The dynamin-like GTPase genes Myxovirus 1 (Mx1) and Mx2 defend mammals against a broad range of viral infections. Loss of Mx1 function in human and mice enhances infectivity by multiple RNA and DNA viruses, including orthomyxoviruses (influenza A), paramyxoviruses (measles), and hepadnaviruses (hepatitis B), whereas loss of Mx2 function leads to decreased resistance to HIV-1 and other viruses. Here we show that both Mx1 and Mx2 have been rendered nonfunctional in Odontoceti cetaceans (toothed whales, including dolphins and orcas). We discovered multiple exon deletions, frameshift mutations, premature stop codons, and transcriptional evidence of decay in the coding sequence of both Mx1 and Mx2 in four species of Odontocetes. We trace the likely loss event for both proteins to soon after the divergence of Odontocetes and Mystocetes (baleen whales) ∼33-37 Mya. Our data raise intriguing questions as to what drove the loss of both Mx1 and Mx2 genes in the Odontoceti lineage, a double loss seen in none of 56 other mammalian genomes, and suggests a hitherto unappreciated fundamental genetic difference in the way these magnificent mammals respond to viral infections.


Asunto(s)
Proteínas de Resistencia a Mixovirus/genética , Isoformas de Proteínas/genética , Ballenas/metabolismo , Animales , Humanos , Filogenia , Ballenas/clasificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA