Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Genome Med ; 15(1): 35, 2023 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-37165454

RESUMEN

BACKGROUND: High sequence identity between segmental duplications (SDs) can facilitate copy number variants (CNVs) via non-allelic homologous recombination (NAHR). These CNVs are one of the fundamental causes of genomic disorders such as the 3q29 deletion syndrome (del3q29S). There are 21 protein-coding genes lost or gained as a result of such recurrent 1.6-Mbp deletions or duplications, respectively, in the 3q29 locus. While NAHR plays a role in CNV occurrence, the factors that increase the risk of NAHR at this particular locus are not well understood. METHODS: We employed an optical genome mapping technique to characterize the 3q29 locus in 161 unaffected individuals, 16 probands with del3q29S and their parents, and 2 probands with the 3q29 duplication syndrome (dup3q29S). Long-read sequencing-based haplotype resolved de novo assemblies from 44 unaffected individuals, and 1 trio was used for orthogonal validation of haplotypes and deletion breakpoints. RESULTS: In total, we discovered 34 haplotypes, of which 19 were novel haplotypes. Among these 19 novel haplotypes, 18 were detected in unaffected individuals, while 1 novel haplotype was detected on the parent-of-origin chromosome of a proband with the del3q29S. Phased assemblies from 44 unaffected individuals enabled the orthogonal validation of 20 haplotypes. In 89% (16/18) of the probands, breakpoints were confined to paralogous copies of a 20-kbp segment within the 3q29 SDs. In one del3q29S proband, the breakpoint was confined to a 374-bp region using long-read sequencing. Furthermore, we categorized del3q29S cases into three classes and dup3q29S cases into two classes based on breakpoints. Finally, we found no evidence of inversions in parent-of-origin chromosomes. CONCLUSIONS: We have generated the most comprehensive haplotype map for the 3q29 locus using unaffected individuals, probands with del3q29S or dup3q29S, and available parents, and also determined the deletion breakpoint to be within a 374-bp region in one proband with del3q29S. These results should provide a better understanding of the underlying genetic architecture that contributes to the etiology of del3q29S and dup3q29S.


Asunto(s)
Genómica , Duplicaciones Segmentarias en el Genoma , Humanos , Mapeo Cromosómico , Síndrome , Haplotipos , Variaciones en el Número de Copia de ADN
2.
Front Neurol ; 13: 918022, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35911904

RESUMEN

We report the genetic analysis of two consanguineous pedigrees of Pakistani ancestry in which two siblings in each family exhibited developmental delay, epilepsy, intellectual disability and aggressive behavior. Whole-genome sequencing was performed in Family 1, and we identified ~80,000 variants located in regions of homozygosity. Of these, 615 variants had a minor allele frequency ≤ 0.001, and 21 variants had CADD scores ≥ 15. Four homozygous exonic variants were identified in both affected siblings: PDZD7 (c.1348_1350delGAG, p.Glu450del), ALG6 (c.1033G>C, p.Glu345Gln), RBM20 (c.1587C>G, p.Ser529Arg), and CNTNAP2 (c.785G>A, p.Gly228Arg). Sanger sequencing revealed co-segregation of the PDZD7, RBM20, and CNTNAP2 variants with disease in Family 1. Pathogenic variants in PDZD7 and RBM20 are associated with autosomal recessive non-syndromic hearing loss and autosomal dominant dilated cardiomyopathy, respectively, suggesting that these variants are unlikely likely to contribute to the clinical presentation. Gene panel analysis was performed on the two affected siblings in Family 2, and they were found to also be homozygous for the p.Gly228Arg CNTNAP2 variant. Together these families provide a LOD score 2.9 toward p.Gly228Arg CNTNAP2 being a completely penetrant recessive cause of this disease. The clinical presentation of the affected siblings in both families is also consistent with previous reports from individuals with homozygous CNTNAP2 variants where at least one allele was a nonsense variant, frameshift or small deletion. Our data suggests that homozygous CNTNAP2 missense variants can also contribute to disease, thereby expanding the genetic landscape of CNTNAP2 dysfunction.

3.
Am J Hum Genet ; 109(8): 1500-1519, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35931052

RESUMEN

Identifying causative gene(s) within disease-associated large genomic regions of copy-number variants (CNVs) is challenging. Here, by targeted sequencing of genes within schizophrenia (SZ)-associated CNVs in 1,779 SZ cases and 1,418 controls, we identified three rare putative loss-of-function (LoF) mutations in OTU deubiquitinase 7A (OTUD7A) within the 15q13.3 deletion in cases but none in controls. To tie OTUD7A LoF with any SZ-relevant cellular phenotypes, we modeled the OTUD7A LoF mutation, rs757148409, in human induced pluripotent stem cell (hiPSC)-derived induced excitatory neurons (iNs) by CRISPR-Cas9 engineering. The mutant iNs showed a ∼50% decrease in OTUD7A expression without undergoing nonsense-mediated mRNA decay. The mutant iNs also exhibited marked reduction of dendritic complexity, density of synaptic proteins GluA1 and PSD-95, and neuronal network activity. Congruent with the neuronal phenotypes in mutant iNs, our transcriptomic analysis showed that the set of OTUD7A LoF-downregulated genes was enriched for those relating to synapse development and function and was associated with SZ and other neuropsychiatric disorders. These results suggest that OTUD7A LoF impairs synapse development and neuronal function in human neurons, providing mechanistic insight into the possible role of OTUD7A in driving neuropsychiatric phenotypes associated with the 15q13.3 deletion.


Asunto(s)
Células Madre Pluripotentes Inducidas , Esquizofrenia , Variaciones en el Número de Copia de ADN , Humanos , Neuronas , Esquizofrenia/metabolismo , Sinapsis/metabolismo
4.
Proc Natl Acad Sci U S A ; 119(22): e2118124119, 2022 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-35617426

RESUMEN

Fragile X­associated tremor/ataxia syndrome (FXTAS) is a debilitating late-onset neurodegenerative disease in premutation carriers of the expanded CGG repeat in FMR1 that presents with a spectrum of neurological manifestations, such as gait ataxia, intention tremor, and parkinsonism [P. J. Hagerman, R. J. Hagerman, Ann. N. Y. Acad. Sci. 1338, 58­70 (2015); S. Jacquemont et al., JAMA 291, 460­469 (2004)]. Here, we performed whole-genome sequencing (WGS) on male premutation carriers (CGG55­200) and prioritized candidate variants to screen for candidate genetic modifiers using a Drosophila model of FXTAS. We found 18 genes that genetically modulate CGG-associated neurotoxicity in Drosophila, such as Prosbeta5 (PSMB5), pAbp (PABPC1L), e(y)1 (TAF9), and CG14231 (OSGEPL1). Among them, knockdown of Prosbeta5 (PSMB5) suppressed CGG-associated neurodegeneration in the fly as well as in N2A cells. Interestingly, an expression quantitative trait locus variant in PSMB5, PSMB5rs11543947-A, was found to be associated with decreased expression of PSMB5 and delayed onset of FXTAS in human FMR1 premutation carriers. Finally, we demonstrate evidence that PSMB5 knockdown results in suppression of CGG neurotoxicity via both the RAN translation and RNA-mediated toxicity mechanisms, thereby presenting a therapeutic strategy for FXTAS.


Asunto(s)
Ataxia , Síndrome del Cromosoma X Frágil , Complejo de la Endopetidasa Proteasomal , Temblor , Animales , Ataxia/genética , Modelos Animales de Enfermedad , Drosophila melanogaster , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Síndrome del Cromosoma X Frágil/genética , Humanos , Masculino , Complejo de la Endopetidasa Proteasomal/genética , Temblor/genética
5.
Transl Psychiatry ; 12(1): 66, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177588

RESUMEN

The 1.6 Mb 3q29 deletion is associated with developmental and psychiatric phenotypes, including a 40-fold increased risk for schizophrenia. Reduced birth weight and a high prevalence of feeding disorders in patients suggest underlying metabolic dysregulation. We investigated 3q29 deletion-induced metabolic changes using our previously generated heterozygous B6.Del16+/Bdh1-Tfrc mouse model. Animals were provided either standard chow (STD) or high-fat diet (HFD). Growth curves were performed on HFD mice to assess weight change (n = 30-50/group). Indirect calorimetry and untargeted metabolomics were performed on STD and HFD mice to evaluate metabolic phenotypes (n = 8-14/group). A behavioral battery was performed on STD and HFD mice to assess behavior change after the HFD challenge (n = 5-13/group). We found that B6.Del16+/Bdh1-Tfrc animals preferentially use dietary lipids as an energy source. Untargeted metabolomics of liver tissue showed a strong sex-dependent effect of the 3q29 deletion on fat metabolism. A HFD partially rescued the 3q29 deletion-associated weight deficit in females, but not males. Untargeted metabolomics of liver tissue after HFD revealed persistent fat metabolism alterations in females. The HFD did not affect B6.Del16+/Bdh1-Tfrc behavioral phenotypes, suggesting that 3q29 deletion-associated metabolic and behavioral outcomes are uncoupled. Our data suggest that dietary interventions to improve weight phenotypes in 3q29 deletion syndrome patients are unlikely to exacerbate behavioral manifestations. Our study also highlights the importance of assessing sex in metabolic studies and suggests that mechanisms underlying 3q29 deletion-associated metabolic phenotypes are sex-specific.


Asunto(s)
Discapacidad Intelectual , Esquizofrenia , Animales , Niño , Deleción Cromosómica , Discapacidades del Desarrollo/genética , Dieta Alta en Grasa , Femenino , Humanos , Discapacidad Intelectual/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Fenotipo , Esquizofrenia/complicaciones , Esquizofrenia/genética
7.
BMC Med Genomics ; 14(1): 154, 2021 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107974

RESUMEN

BACKGROUND: Structural rearrangements of the genome, which generally occur during meiosis and result in large-scale (> 1 kb) copy number variants (CNV; deletions or duplications ≥ 1 kb), underlie genomic disorders. Recurrent pathogenic CNVs harbor similar breakpoints in multiple unrelated individuals and are primarily formed via non-allelic homologous recombination (NAHR). Several pathogenic NAHR-mediated recurrent CNV loci demonstrate biases for parental origin of de novo CNVs. However, the mechanism underlying these biases is not well understood. METHODS: We performed a systematic, comprehensive literature search to curate parent of origin data for multiple pathogenic CNV loci. Using a regression framework, we assessed the relationship between parental CNV origin and the male to female recombination rate ratio. RESULTS: We demonstrate significant association between sex-specific differences in meiotic recombination and parental origin biases at these loci (p = 1.07 × 10-14). CONCLUSIONS: Our results suggest that parental origin of CNVs is largely influenced by sex-specific recombination rates and highlight the need to consider these differences when investigating mechanisms that cause structural variation.


Asunto(s)
Genómica
8.
J Biomol Tech ; 32(2): 74-82, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34121934

RESUMEN

Across the United States, the number of staff scientists (master's- or doctoral-level professionals working in nonfaculty roles) has grown by 35% since 2010, and they play an increasingly important role in research efforts. However, few targeted resources are available, which potentially limits the effectiveness of this group. Launched in 2016, the staff scientist path at Emory has tripled in size over 4 y to 138 staff. The present case study evaluated the perceptions of staff scientists related to onboarding experiences and professional development needs, including those needs arising from coronavirus disease 2019 (COVID-19) impacts in the workplace. A survey of Emory staff scientists was conducted from May to June 2019 as part of a program evaluation initiative to assess perceptions of onboarding and professional development opportunities. Interviews with a subset of scientists informed the survey development and identified COVID-19-related impacts on daily work. Results indicated the need for targeted orientation resources specific to staff scientists, accurate and timely information and resources to support scientists' supervisors, and professional development for scientists in leadership and management-related skills. Remote work associated with COVID-19 accentuated the need for managerial skills, including team development in digital work environments. Findings from this case study can inform policies and practices at Emory and other institutions that employ a similar staff scientist model.


Asunto(s)
COVID-19/epidemiología , Médicos/estadística & datos numéricos , SARS-CoV-2/genética , Lugar de Trabajo , COVID-19/genética , COVID-19/virología , Movilidad Laboral , Femenino , Personal de Salud , Humanos , Masculino , Médicos/psicología , SARS-CoV-2/patogenicidad
9.
Inflamm Bowel Dis ; 27(12): 1889-1900, 2021 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-34002220

RESUMEN

BACKGROUND: The gut and oral microbiome have independently been shown to be associated with inflammatory bowel disease (IBD). However, it is not known to what extent gut and oral microbial disease markers converge in terms of their composition in IBD. Further, the spatial and temporal variation within the oral microenvironments of IBD remain to be elucidated. PATIENTS AND METHODS: We used a prospectively recruited cohort of patients with IBD (n = 47) and unrelated healthy control patients (n = 18) to examine the spatial and temporal distribution of microbiota within the various oral microenvironments, represented by saliva, tongue, buccal mucosa, and plaque, and compared them with stool. Microbiome characterization was performed using 16S rRNA gene sequencing. RESULTS: The oral microbiome displayed IBD-associated dysbiosis, in a site- and taxa-specific manner. Plaque samples depicted a relatively severe degree of dysbiosis, and the disease-associated dysbiotic bacterial groups were predominantly the members of the phylum Firmicutes. Our 16S rRNA gene analyses show that oral microbiota can distinguish patients with IBD from healthy control patients, with salivary microbiota performing the best, closely matched by stool and other oral sites. Longitudinal profiles of microbial composition suggest that some taxa are more consistently perturbed than others, preferentially in a site-dependent fashion. CONCLUSIONS: Collectively, these data indicate the potential of using oral microbial profiles in screening and monitoring patients with IBD. Furthermore, these results support the importance of spatial and longitudinal microbiome sampling to interpret disease-associated dysbiotic states and eventually to gain insights into disease pathogenesis.


Asunto(s)
Enfermedades Inflamatorias del Intestino , Microbiota , Boca/microbiología , Disbiosis/diagnóstico , Heces/microbiología , Humanos , Enfermedades Inflamatorias del Intestino/clasificación , Enfermedades Inflamatorias del Intestino/microbiología , ARN Ribosómico 16S/genética
10.
Fertil Steril ; 116(3): 843-854, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34016428

RESUMEN

OBJECTIVE: To identify modifying genes that explains the risk of fragile X-associated primary ovarian insufficiency (FXPOI). DESIGN: Gene-based, case/control association study, followed by a functional screen of highly ranked genes using a Drosophila model. SETTING: Participants were recruited from academic and clinical settings. PATIENT(S): Women with a premutation (PM) who experienced FXPOI at the age of 35 years or younger (n = 63) and women with a PM who experienced menopause at the age of 50 years or older (n = 51) provided clinical information and a deoxyribonucleic acid sample for whole genome sequencing. The functional screen was on the basis of Drosophila TRiP lines. INTERVENTION(S): Clinical information and a DNA sample were collected for whole genome sequencing. MAIN OUTCOME MEASURES: A polygenic risk score derived from common variants associated with natural age at menopause was calculated and associated with the risk of FXPOI. Genes associated with the risk of FXPOI were identified on the basis of the P-value from gene-based association test and an altered level of fecundity when knocked down in the Drosophila PM model. RESULTS: The polygenic risk score on the basis of common variants associated with natural age at menopause explained approximately 8% of the variance in the risk of FXPOI. Further, SUMO1 and KRR1 were identified as possible modifying genes associated with the risk of FXPOI on the basis of an untargeted gene analysis of rare variants. CONCLUSIONS: In addition to the large genetic effect of a PM on ovarian function, the additive effects of common variants associated with natural age at menopause and the effect of rare modifying variants appear to play a role in FXPOI risk.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Menopausia/genética , Mutación , Ovario/fisiopatología , Insuficiencia Ovárica Primaria/genética , Adulto , Factores de Edad , Animales , Animales Modificados Genéticamente , Estudios de Casos y Controles , Drosophila melanogaster/genética , Femenino , Fertilidad/genética , Antecedentes Genéticos , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Persona de Mediana Edad , Fenotipo , Insuficiencia Ovárica Primaria/diagnóstico , Insuficiencia Ovárica Primaria/fisiopatología , Medición de Riesgo , Factores de Riesgo
11.
J Biomol Tech ; 32(1): 36-41, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-34025222

RESUMEN

Force multipliers are attributes of an organization that enable the successful completion of multiple essential missions. Core facilities play a critical role in the research enterprise and can be organized as force multipliers. Conceiving of cores in this way influences their organization, funding, and research impact. To function as a force multiplier for the research enterprise, core facilities need to do more than efficiently provide services for investigators and generate revenue to recover their service costs: they must be aligned with the strategic objectives of a research university. When core facilities are organized in this way, they can facilitate recruitment of faculty and trainees; serve to retain talented faculty; drive, acquire, and maintain cutting-edge research platforms; and promote interaction and collaboration across the institution. Most importantly, cores accelerate the discovery and sharing of knowledge that are the foundation of a modern research university. This idea has been systematically implemented through the Emory Integrated Core Facilities (cores.emory.edu), which include 16 distinct core facilities and the Division of Animal Resources. Force multiplier core facilities can significantly contribute to the many essential missions necessary for the success of the research enterprise at research universities.


Asunto(s)
Investigadores , Universidades , Humanos
12.
Sci Rep ; 11(1): 8523, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33875724

RESUMEN

Lesch-Nyhan disease (LND) is an inherited disorder caused by pathogenic variants in the HPRT1 gene, which encodes the purine recycling enzyme hypoxanthine-guanine phosphoribosyltransferase (HGprt). We generated 6 induced pluripotent stem cell (iPSC) lines from 3 individuals with LND, along with 6 control lines from 3 normal individuals. All 12 lines had the characteristics of pluripotent stem cells, as assessed by immunostaining for pluripotency markers, expression of pluripotency genes, and differentiation into the 3 primary germ cell layers. Gene expression profiling with RNAseq demonstrated significant heterogeneity among the lines. Despite this heterogeneity, several anticipated abnormalities were readily detectable across all LND lines, including reduced HPRT1 mRNA. Several unexpected abnormalities were also consistently detectable across the LND lines, including decreases in FAR2P1 and increases in RNF39. Shotgun proteomics also demonstrated several expected abnormalities in the LND lines, such as absence of HGprt protein. The proteomics study also revealed several unexpected abnormalities across the LND lines, including increases in GNAO1 decreases in NSE4A. There was a good but partial correlation between abnormalities revealed by the RNAseq and proteomics methods. Finally, functional studies demonstrated LND lines had no HGprt enzyme activity and resistance to the toxic pro-drug 6-thioguanine. Intracellular purines in the LND lines were normal, but they did not recycle hypoxanthine. These cells provide a novel resource to reveal insights into the relevance of heterogeneity among iPSC lines and applications for modeling LND.


Asunto(s)
Células Madre Pluripotentes Inducidas/citología , Síndrome de Lesch-Nyhan/patología , Adolescente , Adulto , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Niño , Perfilación de la Expresión Génica/métodos , Humanos , Hipoxantina Fosforribosiltransferasa/genética , Síndrome de Lesch-Nyhan/genética , Masculino , Purinas/metabolismo , ARN Mensajero/genética , Adulto Joven
13.
J Biomol Tech ; 2021 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-33584149

RESUMEN

Force multipliers are attributes of an organization that enable the successful completion of multiple essential missions. Core facilities play a critical role in the research enterprise and can be organized as force multipliers. Conceiving of cores in this way influences their organization, funding, and research impact. To function as a force multiplier for the research enterprise, core facilities need to do more than efficiently provide services for investigators and generate revenue to recover their service costs: they must be aligned with the strategic objectives of a research university. When core facilities are organized in this way, they can facilitate recruitment of faculty and trainees; serve to retain talented faculty; drive, acquire, and maintain cutting-edge research platforms; and promote interaction and collaboration across the institution. Most importantly, cores accelerate the discovery and sharing of knowledge that are the foundation of a modern research university. This idea has been systematically implemented through the Emory Integrated Core Facilities (cores.emory.edu), which include 16 distinct core facilities and the Division of Animal Resources. Force multiplier core facilities can significantly contribute to the many essential missions necessary for the success of the research enterprise at research universities.

14.
Mol Psychiatry ; 26(8): 4496-4510, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32015465

RESUMEN

Schizophrenia occurs in about one in four individuals with 22q11.2 deletion syndrome (22q11.2DS). The aim of this International Brain and Behavior 22q11.2DS Consortium (IBBC) study was to identify genetic factors that contribute to schizophrenia, in addition to the ~20-fold increased risk conveyed by the 22q11.2 deletion. Using whole-genome sequencing data from 519 unrelated individuals with 22q11.2DS, we conducted genome-wide comparisons of common and rare variants between those with schizophrenia and those with no psychotic disorder at age ≥25 years. Available microarray data enabled direct comparison of polygenic risk for schizophrenia between 22q11.2DS and independent population samples with no 22q11.2 deletion, with and without schizophrenia (total n = 35,182). Polygenic risk for schizophrenia within 22q11.2DS was significantly greater for those with schizophrenia (padj = 6.73 × 10-6). Novel reciprocal case-control comparisons between the 22q11.2DS and population-based cohorts showed that polygenic risk score was significantly greater in individuals with psychotic illness, regardless of the presence of the 22q11.2 deletion. Within the 22q11.2DS cohort, results of gene-set analyses showed some support for rare variants affecting synaptic genes. No common or rare variants within the 22q11.2 deletion region were significantly associated with schizophrenia. These findings suggest that in addition to the deletion conferring a greatly increased risk to schizophrenia, the risk is higher when the 22q11.2 deletion and common polygenic risk factors that contribute to schizophrenia in the general population are both present.


Asunto(s)
Síndrome de DiGeorge , Trastornos Psicóticos , Esquizofrenia , Adulto , Estudios de Casos y Controles , Estudios de Cohortes , Síndrome de DiGeorge/genética , Humanos , Esquizofrenia/genética
15.
Sci Rep ; 10(1): 18051, 2020 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-33093519

RESUMEN

Atrioventricular septal defects (AVSD) are a severe congenital heart defect present in individuals with Down syndrome (DS) at a > 2000-fold increased prevalence compared to the general population. This study aimed to identify risk-associated genes and pathways and to examine a potential polygenic contribution to AVSD in DS. We analyzed a total cohort of 702 individuals with DS with or without AVSD, with genomic data from whole exome sequencing, whole genome sequencing, and/or array-based imputation. We utilized sequence kernel association testing and polygenic risk score (PRS) methods to examine rare and common variants. Our findings suggest that the Notch pathway, particularly NOTCH4, as well as genes involved in the ciliome including CEP290 may play a role in AVSD in DS. These pathways have also been implicated in DS-associated AVSD in prior studies. A polygenic component for AVSD in DS has not been examined previously. Using weights based on the largest genome-wide association study of congenital heart defects available (2594 cases and 5159 controls; all general population samples), we found PRS to be associated with AVSD with odds ratios ranging from 1.2 to 1.3 per standard deviation increase in PRS and corresponding liability r2 values of approximately 1%, suggesting at least a small polygenic contribution to DS-associated AVSD. Future studies with larger sample sizes will improve identification and quantification of genetic contributions to AVSD in DS.


Asunto(s)
Antígenos de Neoplasias , Proteínas de Ciclo Celular , Proteínas del Citoesqueleto , Síndrome de Down/genética , Estudio de Asociación del Genoma Completo , Defectos de los Tabiques Cardíacos/genética , Receptor Notch4 , Estudios de Cohortes , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Riesgo , Secuenciación Completa del Genoma
16.
PLoS One ; 15(9): e0238497, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32986729

RESUMEN

Human papilloma virus (HPV) causes a subset of head and neck squamous cell carcinomas (HNSCC) of the oropharynx. We combined targeted DNA- and genome-wide RNA-sequencing to identify genetic variants and gene expression signatures respectively from patients with HNSCC including oropharyngeal squamous cell carcinomas (OPSCC). DNA and RNA were purified from 35- formalin fixed and paraffin embedded (FFPE) HNSCC tumor samples. Immuno-histochemical evaluation of tumors was performed to determine the expression levels of p16INK4A and classified tumor samples either p16+ or p16-. Using ClearSeq Comprehensive Cancer panel, we examined the distribution of somatic mutations. Somatic single-nucleotide variants (SNV) were called using GATK-Mutect2 ("tumor-only" mode) approach. Using RNA-seq, we identified a catalog of 1,044 and 8 genes as significantly expressed between p16+ and p16-, respectively at FDR 0.05 (5%) and 0.1 (10%). The clinicopathological characteristics of the patients including anatomical site, smoking and survival were analyzed when comparing p16+ and p16- tumors. The majority of tumors (65%) were p16+. Population sequence variant databases, including gnomAD, ExAC, COSMIC and dbSNP, were used to identify the mutational landscape of somatic sequence variants within sequenced genes. Hierarchical clustering of The Cancer Genome Atlas (TCGA) samples based on HPV-status was observed using differentially expressed genes. Using RNA-seq in parallel with targeted DNA-seq, we identified mutational and gene expression signatures characteristic of p16+ and p16- HNSCC. Our gene signatures are consistent with previously published data including TCGA and support the need to further explore the biologic relevance of these alterations in HNSCC.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Adulto , Anciano , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , ADN Viral/genética , Manejo de Datos , Bases de Datos de Ácidos Nucleicos , Pruebas Diagnósticas de Rutina , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/virología , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neoplasias Orofaríngeas/genética , Neoplasias Orofaríngeas/metabolismo , Neoplasias Orofaríngeas/virología , Papillomaviridae/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/virología , Transcriptoma
17.
Am J Epidemiol ; 189(12): 1610-1622, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32639515

RESUMEN

Modern epidemiologic studies permit investigation of the complex pathways that mediate effects of social, behavioral, and molecular factors on health outcomes. Conventional analytical approaches struggle with high-dimensional data, leading to high likelihoods of both false-positive and false-negative inferences. Herein, we describe a novel Bayesian pathway analysis approach, the algorithm for learning pathway structure (ALPS), which addresses key limitations in existing approaches to complex data analysis. ALPS uses prior information about pathways in concert with empirical data to identify and quantify complex interactions within networks of factors that mediate an association between an exposure and an outcome. We illustrate ALPS through application to a complex gene-drug interaction analysis in the Predictors of Breast Cancer Recurrence (ProBe CaRe) Study, a Danish cohort study of premenopausal breast cancer patients (2002-2011), for which conventional analyses severely limit the quality of inference.


Asunto(s)
Algoritmos , Teorema de Bayes , Resistencia a Antineoplásicos/genética , Pruebas de Farmacogenómica , Antineoplásicos Hormonales/metabolismo , Antineoplásicos Hormonales/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Femenino , Humanos , Tamoxifeno/metabolismo , Tamoxifeno/uso terapéutico
18.
Am J Med Genet A ; 182(5): 1152-1166, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32154651

RESUMEN

3q29 duplication syndrome (3q29dup) is a rare genomic disorder caused by a 1.6 Mb duplication (GRCh38 chr3:195,998,000-197,623,000). Case reports indicate the 3q29dup is likely to be pathogenic, but the full range of manifestations is not well understood. We used the 3q29 registry (https://3q29.com) to ascertain 31 individuals with 3q29dup, the largest cohort ever surveyed in a systematic way. For comparison, we ascertained 117 individuals with the reciprocal 3q29 deletion and 64 typically developing controls. We used a custom medical and demographic questionnaire to assess physical and developmental phenotypes, and two standardized instruments, the Social Responsiveness Scale and Child Behavior Checklist/Adult Behavior Checklist, to assess social disability. Participants with 3q29dup report a high rate of problems in the first year of life (80.6%), including feeding problems (55%), failure to gain weight (42%), hypotonia (39%), and respiratory distress (29%). In early childhood, learning problems (71.0%) and seizures (25.8%) are common. Additionally, the rate of self-reported autism spectrum disorder diagnoses (39%) is substantially elevated compared to the general population, suggesting that the 3q29 duplication may be an autism susceptibility locus. This is the most comprehensive description of 3q29dup to date. Our findings can be used to develop evidence-based strategies for early intervention and management of 3q29dup.


Asunto(s)
Duplicación Cromosómica/genética , Cromosomas Humanos Par 3/genética , Discapacidades del Desarrollo/genética , Discapacidad Intelectual/genética , Adolescente , Adulto , Trastorno del Espectro Autista/epidemiología , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Niño , Preescolar , Deleción Cromosómica , Discapacidades del Desarrollo/epidemiología , Discapacidades del Desarrollo/fisiopatología , Femenino , Humanos , Discapacidad Intelectual/fisiopatología , Masculino , Persona de Mediana Edad , Fenotipo , Sistema de Registros , Adulto Joven
19.
Cell ; 180(3): 568-584.e23, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31981491

RESUMEN

We present the largest exome sequencing study of autism spectrum disorder (ASD) to date (n = 35,584 total samples, 11,986 with ASD). Using an enhanced analytical framework to integrate de novo and case-control rare variation, we identify 102 risk genes at a false discovery rate of 0.1 or less. Of these genes, 49 show higher frequencies of disruptive de novo variants in individuals ascertained to have severe neurodevelopmental delay, whereas 53 show higher frequencies in individuals ascertained to have ASD; comparing ASD cases with mutations in these groups reveals phenotypic differences. Expressed early in brain development, most risk genes have roles in regulation of gene expression or neuronal communication (i.e., mutations effect neurodevelopmental and neurophysiological changes), and 13 fall within loci recurrently hit by copy number variants. In cells from the human cortex, expression of risk genes is enriched in excitatory and inhibitory neuronal lineages, consistent with multiple paths to an excitatory-inhibitory imbalance underlying ASD.


Asunto(s)
Trastorno Autístico/genética , Corteza Cerebral/crecimiento & desarrollo , Secuenciación del Exoma/métodos , Regulación del Desarrollo de la Expresión Génica , Neurobiología/métodos , Estudios de Casos y Controles , Linaje de la Célula , Estudios de Cohortes , Exoma , Femenino , Frecuencia de los Genes , Predisposición Genética a la Enfermedad , Humanos , Masculino , Mutación Missense , Neuronas/metabolismo , Fenotipo , Factores Sexuales , Análisis de la Célula Individual/métodos
20.
Cancer Epidemiol Biomarkers Prev ; 29(3): 582-590, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31932415

RESUMEN

BACKGROUND: Tamoxifen and its metabolites compete with estrogen to occupy the estrogen receptor. The conventional dose of adjuvant tamoxifen overwhelms estrogen in this competition, reducing breast cancer recurrence risk by nearly half. Phase I metabolism generates active tamoxifen metabolites, and phase II metabolism deactivates them. No earlier pharmacogenetic study has comprehensively evaluated the metabolism and transport pathways, and no earlier study has included a large population of premenopausal women. METHODS: We completed a cohort study of 5,959 Danish nonmetastatic premenopausal breast cancer patients, in whom 938 recurrences occurred, and a case-control study of 541 recurrent cases in a cohort of Danish predominantly postmenopausal breast cancer patients, all followed for 10 years. We collected formalin-fixed paraffin-embedded tumor blocks and genotyped 32 variants in 15 genes involved in tamoxifen metabolism or transport. We estimated conventional associations for each variant and used prior information about the tamoxifen metabolic path to evaluate the importance of metabolic and transporter pathways. RESULTS: No individual variant was notably associated with risk of recurrence in either study population. Both studies showed weak evidence of the importance of phase I metabolism in the clinical response to adjuvant tamoxifen therapy. CONCLUSIONS: Consistent with prior knowledge, our results support the role of phase I metabolic capacity in clinical response to tamoxifen. Nonetheless, no individual variant substantially explained the modest phase I effect on tamoxifen response. IMPACT: These results are consistent with guidelines recommending against genotype-guided prescribing of tamoxifen, and for the first time provide evidence supporting these guidelines in premenopausal women.


Asunto(s)
Antineoplásicos Hormonales/farmacología , Biomarcadores de Tumor/genética , Neoplasias de la Mama/tratamiento farmacológico , Recurrencia Local de Neoplasia/epidemiología , Tamoxifeno/farmacología , Adulto , Antineoplásicos Hormonales/uso terapéutico , Biomarcadores de Tumor/metabolismo , Mama/patología , Mama/cirugía , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Estudios de Casos y Controles , Quimioterapia Adyuvante/métodos , Conjuntos de Datos como Asunto , Dinamarca , Femenino , Estudios de Seguimiento , Técnicas de Genotipaje , Humanos , Mastectomía , Redes y Vías Metabólicas/genética , Persona de Mediana Edad , Recurrencia Local de Neoplasia/genética , Recurrencia Local de Neoplasia/patología , Recurrencia Local de Neoplasia/prevención & control , Pruebas de Farmacogenómica , Variantes Farmacogenómicas , Sistema de Registros/estadística & datos numéricos , Tamoxifeno/uso terapéutico , Resultado del Tratamiento
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA