Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Blood ; 142(2): 172-184, 2023 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-37001051

RESUMEN

Trisomy 21, the genetic cause of Down syndrome (DS), is the most common congenital chromosomal anomaly. It is associated with a 20-fold increased risk of acute lymphoblastic leukemia (ALL) during childhood and results in distinctive leukemia biology. To comprehensively define the genomic landscape of DS-ALL, we performed whole-genome sequencing and whole-transcriptome sequencing (RNA-Seq) on 295 cases. Our integrated genomic analyses identified 15 molecular subtypes of DS-ALL, with marked enrichment of CRLF2-r, IGH::IGF2BP1, and C/EBP altered (C/EBPalt) subtypes compared with 2257 non-DS-ALL cases. We observed abnormal activation of the CEBPD, CEBPA, and CEBPE genes in 10.5% of DS-ALL cases via a variety of genomic mechanisms, including chromosomal rearrangements and noncoding mutations leading to enhancer hijacking. A total of 42.3% of C/EBP-activated DS-ALL also have concomitant FLT3 point mutations or insertions/deletions, compared with 4.1% in other subtypes. CEBPD overexpression enhanced the differentiation of mouse hematopoietic progenitor cells into pro-B cells in vitro, particularly in a DS genetic background. Notably, recombination-activating gene-mediated somatic genomic abnormalities were common in DS-ALL, accounting for a median of 27.5% of structural alterations, compared with 7.7% in non-DS-ALL. Unsupervised hierarchical clustering analyses of CRLF2-rearranged DS-ALL identified substantial heterogeneity within this group, with the BCR::ABL1-like subset linked to an inferior event-free survival, even after adjusting for known clinical risk factors. These results provide important insights into the biology of DS-ALL and point to opportunities for targeted therapy and treatment individualization.


Asunto(s)
Síndrome de Down , Leucemia-Linfoma Linfoblástico de Células Precursoras , Animales , Ratones , Síndrome de Down/complicaciones , Síndrome de Down/genética , Mutación , Factores de Riesgo , Genómica , Aberraciones Cromosómicas , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicaciones , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
2.
Leukemia ; 37(3): 518-528, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36658389

RESUMEN

Childhood B-cell acute lymphoblastic leukaemia (B-ALL) is characterised by recurrent genetic abnormalities that drive risk-directed treatment strategies. Using current techniques, accurate detection of such aberrations can be challenging, due to the rapidly expanding list of key genetic abnormalities. Whole genome sequencing (WGS) has the potential to improve genetic testing, but requires comprehensive validation. We performed WGS on 210 childhood B-ALL samples annotated with clinical and genetic data. We devised a molecular classification system to subtype these patients based on identification of key genetic changes in tumour-normal and tumour-only analyses. This approach detected 294 subtype-defining genetic abnormalities in 96% (202/210) patients. Novel genetic variants, including fusions involving genes in the MAP kinase pathway, were identified. WGS results were concordant with standard-of-care methods and whole transcriptome sequencing (WTS). We expanded the catalogue of genetic profiles that reliably classify PAX5alt and ETV6::RUNX1-like subtypes. Our novel bioinformatic pipeline improved detection of DUX4 rearrangements (DUX4-r): a good-risk B-ALL subtype with high survival rates. Overall, we have validated that WGS provides a standalone, reliable genetic test to detect all subtype-defining genetic abnormalities in B-ALL, accurately classifying patients for the risk-directed treatment stratification, while simultaneously performing as a research tool to identify novel disease biomarkers.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Biología Computacional , Pruebas Genéticas , Secuenciación Completa del Genoma
3.
Nat Genet ; 54(9): 1376-1389, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36050548

RESUMEN

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer. Here, using whole-genome, exome and transcriptome sequencing of 2,754 childhood patients with ALL, we find that, despite a generally low mutation burden, ALL cases harbor a median of four putative somatic driver alterations per sample, with 376 putative driver genes identified varying in prevalence across ALL subtypes. Most samples harbor at least one rare gene alteration, including 70 putative cancer driver genes associated with ubiquitination, SUMOylation, noncoding transcripts and other functions. In hyperdiploid B-ALL, chromosomal gains are acquired early and synchronously before ultraviolet-induced mutation. By contrast, ultraviolet-induced mutations precede chromosomal gains in B-ALL cases with intrachromosomal amplification of chromosome 21. We also demonstrate the prognostic significance of genetic alterations within subtypes. Intriguingly, DUX4- and KMT2A-rearranged subtypes separate into CEBPA/FLT3- or NFATC4-expressing subgroups with potential clinical implications. Together, these results deepen understanding of the ALL genomic landscape and associated outcomes.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células Precursoras , Niño , Aberraciones Cromosómicas , Exoma/genética , Genómica , Humanos , Mutación , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética
4.
Adv Exp Med Biol ; 1361: 163-175, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35230688

RESUMEN

Gene fusions play a prominent role in the oncogenesis of many cancers and have been extensively targeted as biomarkers for diagnostic, prognostic, and therapeutic purposes. Detection methods span a number of platforms, including cytogenetics (e.g., FISH), targeted qPCR, and sequencing-based assays. Before the advent of next-generation sequencing (NGS), fusion testing was primarily targeted to specific genome loci, with assays tailored for previously characterized fusion events. The availability of whole genome sequencing (WGS) and whole transcriptome sequencing (RNA-seq) allows for genome-wide screening for the simultaneous detection of both known and novel fusions. RNA-seq, in particular, offers the possibility of rapid turn-around testing with less dedicated sequencing than WGS. This makes it an attractive target for clinical oncology testing, particularly when transcriptome data can be multi-purposed for tumor classification and additional analyses. Despite considerable efforts and substantial progress, however, genome-wide screening for fusions solely based on RNA-seq data remains an ongoing challenge. A host of technical artifacts adversely impact the sensitivity and specificity of existing software tools. In this chapter, the general strategies employed by current fusion software are discussed, and a selection of available fusion detection tools are surveyed. Despite its current limitations, RNA-seq-based fusion detection offers a more comprehensive and efficient strategy as compared to multiple targeted fusion assays. When thoughtfully employed within a wider ecosystem of diagnostic assays and clinical information, RNA-seq fusion detection represents a powerful tool for precision oncology.


Asunto(s)
Neoplasias , Ecosistema , Fusión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Oncología Médica , Neoplasias/diagnóstico , Neoplasias/genética , Medicina de Precisión , RNA-Seq , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Secuenciación del Exoma
5.
Sci Rep ; 11(1): 22213, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34782706

RESUMEN

Rhabdomyosarcomas (RMS) represent a family of aggressive soft tissue sarcomas that present in both children and adults. Pathologic risk stratification for RMS has been based on histologic subtype, with poor outcomes observed in alveolar rhabdomyosarcoma (ARMS) and the adult-type pleomorphic rhabdomyosarcoma (PRMS) compared to embryonal rhabdomyosarcoma (ERMS). Genomic sequencing studies have expanded the spectrum of RMS, with several new molecularly defined entities, including fusion-driven spindle cell/sclerosing rhabdomyosarcoma (SC/SRMS) and MYOD1-mutant SC/SRMS. Comprehensive genomic analysis has previously defined the mutational and copy number spectrum for the more common ERMS and ARMS and revealed corresponding methylation signatures. Comparatively, less is known about epigenetic correlates for the rare SC/SRMS or PRMS histologic subtypes. Herein, we present exome and RNA sequencing, copy number analysis, and methylation profiling of the largest cohort of molecularly characterized RMS samples to date. In addition to ARMS and ERMS, we identify two novel methylation subtypes, one having SC/SRMS histology and defined by MYOD1 p. L122R mutations and the other matching adult-type PRMS. Selected tumors from adolescent patients grouped with the PRMS methylation class, expanding the age range of these rare tumors. Limited follow-up data suggest that pediatric tumors with MYOD1-mutations are associated with an aggressive clinical course.


Asunto(s)
Biomarcadores de Tumor , Metilación de ADN , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Rabdomiosarcoma/diagnóstico , Rabdomiosarcoma/etiología , Adolescente , Adulto , Anciano , Niño , Preescolar , Biología Computacional/métodos , Variaciones en el Número de Copia de ADN , Diagnóstico Diferencial , Susceptibilidad a Enfermedades , Femenino , Humanos , Inmunohistoquímica , Hibridación in Situ , Lactante , Masculino , Persona de Mediana Edad , Mutación , Rabdomiosarcoma/terapia , Secuenciación Completa del Genoma , Adulto Joven
6.
Cancer Discov ; 11(12): 3008-3027, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34301788

RESUMEN

Genomic studies of pediatric cancer have primarily focused on specific tumor types or high-risk disease. Here, we used a three-platform sequencing approach, including whole-genome sequencing (WGS), whole-exome sequencing (WES), and RNA sequencing (RNA-seq), to examine tumor and germline genomes from 309 prospectively identified children with newly diagnosed (85%) or relapsed/refractory (15%) cancers, unselected for tumor type. Eighty-six percent of patients harbored diagnostic (53%), prognostic (57%), therapeutically relevant (25%), and/or cancer-predisposing (18%) variants. Inclusion of WGS enabled detection of activating gene fusions and enhancer hijacks (36% and 8% of tumors, respectively), small intragenic deletions (15% of tumors), and mutational signatures revealing of pathogenic variant effects. Evaluation of paired tumor-normal data revealed relevance to tumor development for 55% of pathogenic germline variants. This study demonstrates the power of a three-platform approach that incorporates WGS to interrogate and interpret the full range of genomic variants across newly diagnosed as well as relapsed/refractory pediatric cancers. SIGNIFICANCE: Pediatric cancers are driven by diverse genomic lesions, and sequencing has proven useful in evaluating high-risk and relapsed/refractory cases. We show that combined WGS, WES, and RNA-seq of tumor and paired normal tissues enables identification and characterization of genetic drivers across the full spectrum of pediatric cancers. This article is highlighted in the In This Issue feature, p. 2945.


Asunto(s)
Neoplasias , Niño , ADN , Humanos , Mutación , Neoplasias/genética , Análisis de Secuencia de ARN , Secuenciación del Exoma
7.
Virchows Arch ; 476(6): 915-920, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31900635

RESUMEN

BCOR internal tandem duplications (ITDs) and rearrangements are implicated in the oncogenesis of a subset of undifferentiated sarcomas. To date, BCOR ITD sarcomas have been exclusively found in non-appendicular infantile soft tissues, whereas BCOR-rearranged sarcomas occur in both bones and soft tissues affecting a wider patient age range. Little is known about patient outcome in BCOR ITD sarcomas. We present a BCOR-expressing, primary bone, undifferentiated sarcoma case involving an adolescent male's left tibia that, unexpectedly, harbored a BCOR ITD instead of a BCOR rearrangement. Furthermore, the patient achieved a partial histologic response after receiving a Ewing sarcoma chemotherapy regimen. Our case expands the clinical spectrum of BCOR ITD sarcomas and suggests that childhood and adult BCOR-expressing sarcomas with an undifferentiated histology should be considered for both BCOR rearrangement and ITD screening. Accurate BCOR mutation identification in undifferentiated sarcomas is essential to define their clinical spectrum and to develop effective management strategies.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Óseas/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Sarcoma/genética , Adolescente , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/patología , Duplicación de Gen , Humanos , Masculino , Sarcoma/diagnóstico por imagen , Sarcoma/tratamiento farmacológico , Sarcoma/patología , Tibia/diagnóstico por imagen , Tibia/patología
8.
Pediatr Blood Cancer ; 67(2): e28047, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31736278

RESUMEN

PURPOSE: To estimate the absolute number of adult survivors of childhood cancer in the U.S. population who carry a pathogenic or likely pathogenic variant in a cancer predisposition gene. METHODS: Using the Surveillance, Epidemiology, and End Results (SEER) Program, we estimated the number of childhood cancer survivors on December 31, 2016 for each childhood cancer diagnosis, multiplied this by the proportion of carriers of pathogenic/likely pathogenic variants in the St. Jude Lifetime Cohort (SJLIFE) study, and projected the resulting number onto the U.S. RESULTS: Based on genome sequence data, 11.8% of 2450 SJLIFE participants carry a pathogenic/likely pathogenic variant in one of 156 cancer predisposition genes. Given this information, we estimate that 21 800 adult survivors of childhood cancer in the United States carry a pathogenic/likely pathogenic variant in one of these genes. The highest estimated absolute number of variant carriers are among survivors of central nervous system tumors (n = 4300), particularly astrocytoma (n = 1800) and other gliomas (n = 1700), acute lymphoblastic leukemia (n = 4300), and retinoblastoma (n = 3500). The most frequently mutated genes are RB1 (n = 3000), NF1 (n = 2300), and BRCA2 (n = 800). CONCLUSION: Given the increasing number of childhood cancer survivors in the United States, clinicians should counsel survivors regarding their potential genetic risk, consider referral for genetic counseling and testing, and, as appropriate, implement syndrome-specific cancer surveillance or risk-reducing measures.


Asunto(s)
Supervivientes de Cáncer/estadística & datos numéricos , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Proteínas de Neoplasias/genética , Neoplasias/mortalidad , Adolescente , Adulto , Anciano , Niño , Preescolar , Estudios de Cohortes , Femenino , Estudios de Seguimiento , Humanos , Incidencia , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Neoplasias/epidemiología , Neoplasias/genética , Pronóstico , Factores de Riesgo , Tasa de Supervivencia , Estados Unidos/epidemiología , Adulto Joven
9.
Genome Res ; 29(9): 1555-1565, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31439692

RESUMEN

Variant interpretation in the era of massively parallel sequencing is challenging. Although many resources and guidelines are available to assist with this task, few integrated end-to-end tools exist. Here, we present the Pediatric Cancer Variant Pathogenicity Information Exchange (PeCanPIE), a web- and cloud-based platform for annotation, identification, and classification of variations in known or putative disease genes. Starting from a set of variants in variant call format (VCF), variants are annotated, ranked by putative pathogenicity, and presented for formal classification using a decision-support interface based on published guidelines from the American College of Medical Genetics and Genomics (ACMG). The system can accept files containing millions of variants and handle single-nucleotide variants (SNVs), simple insertions/deletions (indels), multiple-nucleotide variants (MNVs), and complex substitutions. PeCanPIE has been applied to classify variant pathogenicity in cancer predisposition genes in two large-scale investigations involving >4000 pediatric cancer patients and serves as a repository for the expert-reviewed results. PeCanPIE was originally developed for pediatric cancer but can be easily extended for use for nonpediatric cancers and noncancer genetic diseases. Although PeCanPIE's web-based interface was designed to be accessible to non-bioinformaticians, its back-end pipelines may also be run independently on the cloud, facilitating direct integration and broader adoption. PeCanPIE is publicly available and free for research use.


Asunto(s)
Biología Computacional/métodos , Mutación de Línea Germinal , Neoplasias/genética , Niño , Nube Computacional , Bases de Datos Genéticas , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Interfaz Usuario-Computador
10.
J Clin Oncol ; 36(20): 2078-2087, 2018 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-29847298

RESUMEN

Purpose Childhood cancer survivors are at increased risk of subsequent neoplasms (SNs), but the germline genetic contribution is largely unknown. We assessed the contribution of pathogenic/likely pathogenic (P/LP) mutations in cancer predisposition genes to their SN risk. Patients and Methods Whole-genome sequencing (30-fold) was performed on samples from childhood cancer survivors who were ≥ 5 years since initial cancer diagnosis and participants in the St Jude Lifetime Cohort Study, a retrospective hospital-based study with prospective clinical follow-up. Germline mutations in 60 genes known to be associated with autosomal dominant cancer predisposition syndromes with moderate to high penetrance were classified by their pathogenicity according to the American College of Medical Genetics and Genomics guidelines. Relative rates (RRs) and 95% CIs of SN occurrence by mutation status were estimated using multivariable piecewise exponential regression stratified by radiation exposure. Results Participants were 3,006 survivors (53% male; median age, 35.8 years [range, 7.1 to 69.8 years]; 56% received radiotherapy), 1,120 SNs were diagnosed among 439 survivors (14.6%), and 175 P/LP mutations were identified in 5.8% (95% CI, 5.0% to 6.7%) of survivors. Mutations were associated with significantly increased rates of breast cancer (RR, 13.9; 95% CI, 6.0 to 32.2) and sarcoma (RR, 10.6; 95% CI, 4.3 to 26.3) among irradiated survivors and with increased rates of developing any SN (RR, 4.7; 95% CI, 2.4 to 9.3), breast cancer (RR, 7.7; 95% CI, 2.4 to 24.4), nonmelanoma skin cancer (RR, 11.0; 95% CI, 2.9 to 41.4), and two or more histologically distinct SNs (RR, 18.6; 95% CI, 3.5 to 99.3) among nonirradiated survivors. Conclusion The findings support referral of all survivors for genetic counseling for potential clinical genetic testing, which should be prioritized for nonirradiated survivors with any SN and for those with breast cancer or sarcoma in the field of prior irradiation.


Asunto(s)
Supervivientes de Cáncer/estadística & datos numéricos , Neoplasias Primarias Secundarias/genética , Neoplasias/genética , Adolescente , Adulto , Anciano , Niño , Estudios de Cohortes , Femenino , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal , Humanos , Masculino , Persona de Mediana Edad , Neoplasias/epidemiología , Neoplasias Primarias Secundarias/epidemiología , Estudios Retrospectivos , Riesgo , Estados Unidos/epidemiología , Secuenciación Completa del Genoma , Adulto Joven
11.
BMC Genomics ; 16: 220, 2015 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-25887476

RESUMEN

BACKGROUND: There are over a half a million copies of L1 retroelements in the human genome which are responsible for as much as 0.5% of new human genetic diseases. Most new L1 inserts arise from young source elements that are polymorphic in the human genome. Highly active polymorphic "hot" L1 source elements have been shown to be capable of extremely high levels of mobilization and result in numerous instances of disease. Additionally, hot polymorphic L1s have been described to be highly active within numerous cancer genomes. These hot L1s result in mutagenesis by insertion of new L1 copies elsewhere in the genome, but also have been shown to generate additional full length L1 insertions which are also hot and able to further retrotranspose. Through this mechanism, hot L1s may amplify within a tumor and result in a continued cycle of mutagenesis. RESULTS AND CONCLUSIONS: We have developed a method to detect full-length, polymorphic L1 elements using a targeted next generation sequencing approach, Sequencing Identification and Mapping of Primed L1 Elements (SIMPLE). SIMPLE has 94% sensitivity and detects nearly all full-length L1 elements in a genome. SIMPLE will allow researchers to identify hot mutagenic full-length L1s as potential drivers of genome instability. Using SIMPLE we find that the typical individual has approximately 100 non-reference, polymorphic L1 elements in their genome. These elements are at relatively low population frequencies relative to previously identified polymorphic L1 elements and demonstrate the tremendous diversity in potentially active L1 elements in the human population.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Polimorfismo Genético , Análisis de Secuencia de ADN/métodos , Alelos , Línea Celular , Mapeo Cromosómico , Fibroblastos/metabolismo , Frecuencia de los Genes , Estudios de Asociación Genética , Genoma Humano , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos
12.
Nat Rev Cardiol ; 10(9): 531-47, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23900355

RESUMEN

Remarkable progress has been made in understanding the genetic basis of dilated cardiomyopathy (DCM). Rare variants in >30 genes, some also involved in other cardiomyopathies, muscular dystrophy, or syndromic disease, perturb a diverse set of important myocardial proteins to produce a final DCM phenotype. Large, publicly available datasets have provided the opportunity to evaluate previously identified DCM-causing mutations, and to examine the population frequency of sequence variants similar to those that have been observed to cause DCM. The frequency of these variants, whether associated with dilated or hypertrophic cardiomyopathy, is greater than estimates of disease prevalence. This mismatch might be explained by one or more of the following possibilities: that the penetrance of DCM-causing mutations is lower than previously thought, that some variants are noncausal, that DCM prevalence is higher than previously estimated, or that other more-complex genomics underlie DCM. Reassessment of our assumptions about the complexity of the genomic and phenomic architecture of DCM is warranted. Much about the genomic basis of DCM remains to be investigated, which will require comprehensive genomic studies in much larger cohorts of rigorously phenotyped probands and family members than previously examined.


Asunto(s)
Cardiomiopatía Dilatada/genética , Mutación , Animales , Displasia Ventricular Derecha Arritmogénica/genética , Displasia Ventricular Derecha Arritmogénica/fisiopatología , Cardiomiopatía Dilatada/clasificación , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Hipertrófica Familiar/genética , Cardiomiopatía Hipertrófica Familiar/fisiopatología , Predisposición Genética a la Enfermedad , Genómica/métodos , Herencia , Humanos , Linaje , Fenotipo , Factores de Riesgo , Terminología como Asunto
13.
Genome Res ; 19(4): 545-55, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19273617

RESUMEN

The human genome contains nearly 1.1 million Alu elements comprising roughly 11% of its total DNA content. Alu elements use a copy and paste retrotransposition mechanism that can result in de novo disease insertion alleles. There are nearly 900,000 old Alu elements from subfamilies S and J that appear to be almost completely inactive, and about 200,000 from subfamily Y or younger, which include a few thousand copies of the Ya5 subfamily which makes up the majority of current activity. Given the much higher copy number of the older Alu subfamilies, it is not known why all of the active Alu elements belong to the younger subfamilies. We present a systematic analysis evaluating the observed sequence variation in the different sections of an Alu element on retrotransposition. The length of the longest number of uninterrupted adenines in the A-tail, the degree of A-tail heterogeneity, the length of the 3' unique end after the A-tail and before the RNA polymerase III terminator, and random mutations found in the right monomer all modulate the retrotransposition efficiency. These changes occur over different evolutionary time frames. The combined impact of sequence changes in all of these regions explains why young Alus are currently causing disease through retrotransposition, and the old Alus have lost their ability to retrotranspose. We present a predictive model to evaluate the retrotransposition capability of individual Alu elements and successfully applied it to identify the first putative source element for a disease-causing Alu insertion in a patient with cystic fibrosis.


Asunto(s)
Elementos Alu/genética , Genoma Humano , Polimorfismo Genético , Secuencias Reguladoras de Ácidos Nucleicos/genética , Retroelementos/genética , Secuencia de Bases , Northern Blotting , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Células HeLa , Humanos , Datos de Secuencia Molecular , Mutación/genética , Poli A/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico
14.
Hum Mutat ; 30(2): 204-11, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18726952

RESUMEN

Apert syndrome (AS) is a severe disorder, characterized by craniosynostosis and complex syndactyly of the hands and feet. Two heterozygous gain-of-function substitutions (Ser252Trp and Pro253Arg) in exon IIIa of fibroblast growth factor receptor 2 (FGFR2) are responsible for >98% of cases. Here we describe two novel mutations in FGFR2 in the two patients in whom a mutation had not previously been found in our cohort of 227 AS cases. The first is a 1.93-kb deletion, removing exon IIIc and substantial portions of the flanking introns. This is the first large FGFR2 deletion described in any individual with craniosynostosis. The other mutation is a 5' truncated Alu insertion into exon IIIc. This is the third Alu insertion identified in AS; all have occurred within an interval of only 104 bp, representing an enrichment of over a million-fold compared to the background genomic rate. We show that the inserted Alu element belongs to a small subfamily, not previously known to be mobile, which we term Alu Yk13. Both the deletion and insertion are likely to act by a similar gain-of-function mechanism in which disruption of exon IIIc leads to illegitimate mesenchymal expression of an FGFR2 spliceform containing the alternatively spliced exon IIIb. All the AS-associated Alu insertions have arisen in the paternal germline; we propose that their enrichment in FGFR2 is driven by positive selection of the mutant spermatogonial progenitors, a mechanism analogous to that explaining why the canonical AS nucleotide substitutions also reach exceptionally high levels in sperm.


Asunto(s)
Acrocefalosindactilia/genética , Elementos Alu/genética , Eliminación de Gen , Mutagénesis Insercional/genética , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Acrocefalosindactilia/diagnóstico , Adolescente , Adulto , Emparejamiento Base , Secuencia de Bases , Preescolar , Análisis Mutacional de ADN , Exones/genética , Padre , Genoma Humano/genética , Humanos , Lactante , Masculino , Datos de Secuencia Molecular
15.
Gene ; 390(1-2): 190-8, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17067767

RESUMEN

The human Long Interspersed Element-1 (LINE-1) and the Short Interspersed Element (SINE) Alu comprise 28% of the human genome. They share the same L1-encoded endonuclease for insertion, which recognizes an A+T-rich sequence. Under a simple model of insertion distribution, this nucleotide preference would lead to the prediction that the populations of both elements would be biased towards A+T-rich regions. Genomic L1 elements do show an A+T-rich bias. In contrast, Alu is biased towards G+C-rich regions when compared to the genome average. Several analyses have demonstrated that relatively recent insertions of both elements show less G+C content bias relative to older elements. We have analyzed the repetitive element and G+C composition of more than 100 pre-insertion loci derived from de novo L1 insertions in cultured human cancer cells, which should represent an evolutionarily unbiased set of insertions. An A+T-rich bias is observed in the 50 bp flanking the endonuclease target site, consistent with the known target site for the L1 endonuclease. The L1, Alu, and G+C content of 20 kb of the de novo pre-insertion loci shows a different set of biases than that observed for fixed L1s in the human genome. In contrast to the insertion sites of genomic L1s, the de novo L1 pre-insertion loci are relatively L1-poor, Alu-rich and G+C neutral. Finally, a statistically significant cluster of de novo L1 insertions was localized in the vicinity of the c-myc gene. These results suggest that the initial insertion preference of L1, while A+T-rich in the initial vicinity of the break site, can be influenced by the broader content of the flanking genomic region and have implications for understanding the dynamics of L1 and Alu distributions in the human genome.


Asunto(s)
Elementos de Nucleótido Esparcido Largo , Elementos Alu , Composición de Base , Secuencia de Bases , Mapeo Cromosómico , ADN/química , ADN/genética , Genoma Humano , Células HeLa , Humanos , Modelos Genéticos , Elementos de Nucleótido Esparcido Corto
16.
Nucleic Acids Res ; 34(5): 1512-21, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16554555

RESUMEN

Long interspersed element-1 elements compose on average one-fifth of mammalian genomes. The expression and retrotransposition of L1 is restricted by a number of cellular mechanisms in order to limit their damage in both germ-line and somatic cells. L1 transcription is largely suppressed in most tissues, but L1 mRNA and/or proteins are still detectable in testes, a number of specific somatic cell types, and malignancies. Down-regulation of L1 expression via premature polyadenylation has been found to be a secondary mechanism of limiting L1 expression. We demonstrate that mammalian L1 elements contain numerous functional splice donor and acceptor sites. Efficient usage of some of these sites results in extensive and complex splicing of L1. Several splice variants of both the human and mouse L1 elements undergo retrotransposition. Some of the spliced L1 mRNAs can potentially contribute to expression of open reading frame 2-related products and therefore have implications for the mobility of SINEs even if they are incompetent for L1 retrotransposition. Analysis of the human EST database revealed that L1 elements also participate in splicing events with other genes. Such contribution of functional splice sites by L1 may result in disruption of normal gene expression or formation of alternative mRNA transcripts.


Asunto(s)
Empalme Alternativo , Elementos de Nucleótido Esparcido Largo , Animales , Línea Celular , Regulación de la Expresión Génica , Células HeLa , Humanos , Ratones , Células 3T3 NIH , Poliadenilación , Sitios de Empalme de ARN , ARN Mensajero/metabolismo
17.
Bioessays ; 27(8): 785-94, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16015599

RESUMEN

As is the case with mammals in general, primate genomes are inundated with repetitive sequence. Although much of this repetitive content consists of "molecular fossils" inherited from early mammalian ancestors, a significant portion of this material comprises active mobile element lineages. Despite indications that these elements played a major role in shaping the architecture of the genome, there remain many unanswered questions surrounding the nature of the host-element relationship. Here we review advances in our understanding of the host-mobile element dynamic and its overall impact on primate evolution.


Asunto(s)
Evolución Biológica , Genoma , Empalme Alternativo , Animales , Islas de CpG , Metilación de ADN , Evolución Molecular , Exones , Genoma Humano , Mutación de Línea Germinal , Humanos , Elementos de Nucleótido Esparcido Largo , Modelos Genéticos , Sistemas de Lectura Abierta , Filogenia , Primates , Secuencias Repetitivas de Ácidos Nucleicos , Retroviridae/genética , Elementos de Nucleótido Esparcido Corto
18.
Genome Res ; 12(9): 1333-44, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12213770

RESUMEN

Long and short interspersed elements (LINEs and SINEs) are retroelements that make up almost half of the human genome. L1 and Alu represent the most prolific human LINE and SINE families, respectively. Only a few Alu elements are able to retropose, and the factors determining their retroposition capacity are poorly understood. The data presented in this paper indicate that the length of Alu "A-tails" is one of the principal factors in determining the retropositional capability of an Alu element. The A stretches of the Alu subfamilies analyzed, both old (Alu S and J) and young (Ya5), had a Poisson distribution of A-tail lengths with a mean size of 21 and 26, respectively. In contrast, the A-tails of very recent Alu insertions (disease causing) were all between 40 and 97 bp in length. The L1 elements analyzed displayed a similar tendency, in which the "disease"-associated elements have much longer A-tails (mean of 77) than do the elements even from the young Ta subfamily (mean of 41). Analysis of the draft sequence of the human genome showed that only about 1000 of the over one million Alu elements have tails of 40 or more adenosine residues in length. The presence of these long A stretches shows a strong bias toward the actively amplifying subfamilies, consistent with their playing a major role in the amplification process. Evaluation of the 19 Alu elements retrieved from the draft sequence of the human genome that are identical to the Alu Ya5a2 insert in the NF1 gene showed that only five have tails with 40 or more adenosine residues. Sequence analysis of the loci with the Alu elements containing the longest A-tails (7 of the 19) from the genomes of the NF1 patient and the father revealed that there are at least two loci with A-tails long enough to serve as source elements within our model. Analysis of the A-tail lengths of 12 Ya5a2 elements in diverse human population groups showed substantial variability in both the Alu A-tail length and sequence homogeneity. On the basis of these observations, a model is presented for the role of A-tail length in determining which Alu elements are active.


Asunto(s)
Elementos Alu/genética , Poli A/metabolismo , Secuencias Repetidas Terminales/genética , Región de Flanqueo 3'/genética , Composición de Base/genética , Secuencia de Bases/genética , ADN/genética , Bases de Datos Genéticas , Regulación de la Expresión Génica/genética , Genes de Neurofibromatosis 1 , Marcadores Genéticos , Variación Genética/genética , Genética de Población , Genoma Humano , Humanos , Elementos de Nucleótido Esparcido Largo/genética , Masculino , Datos de Secuencia Molecular
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA