Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Hum Mutat ; 43(11): 1531-1544, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36086952

RESUMEN

Long-read sequencing (LRS) has been around for more than a decade, but widespread adoption of the technology has been slow due to the perceived high error rates and high sequencing cost. This is changing due to the recent advancements to produce highly accurate sequences and the reducing costs. LRS promises significant improvement over short read sequencing in four major areas: (1) better detection of structural variation (2) better resolution of highly repetitive or nonunique regions (3) accurate long-range haplotype phasing and (4) the detection of base modifications natively from the sequencing data. Several successful applications of LRS have demonstrated its ability to resolve molecular diagnoses where short-read sequencing fails to identify a cause. However, the argument for increased diagnostic yield from LRS remains to be validated. Larger cohort studies may be required to establish the realistic boundaries of LRS's clinical utility and analytical validity, as well as the development of standards for clinical applications. We discuss the limitations of the current standard of care, and contrast with the applications and advantages of two major LRS platforms, PacBio and Oxford Nanopore, for molecular diagnostics of constitutional disorders, and present a critical argument about the potential of LRS in diagnostic settings.


Asunto(s)
Secuenciación de Nucleótidos de Alto Rendimiento , Patología Molecular , Humanos , Análisis de Secuencia de ADN
2.
Eur J Med Genet ; 65(9): 104556, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35781022

RESUMEN

KDM5C encodes a demethylase of the histone H3 lysine 4 residue, involved in chromatin regulation and gene expression. Hemizygous KDM5C pathogenic variants cause X-linked intellectual disability of Claes-Jensen type. Because of its mode of inheritance and the low specificity of the clinical phenotype, interpretation of variants can be difficult, hence the need for functional studies and biomarkers specific to this disorder. We present the case of a male patient with intellectual disability, behavioral abnormalities and subtle dysmorphic features, in which genetic investigation identified a hemizygous novel missense KDM5C variant of uncertain significance (VUS), inherited from his asymptomatic mother and present in his paucisymptomatic sister. We assessed the global genomic DNA methylation status from a whole blood sample of the proband. Global DNA methylation profiling specifically identified the recently discovered epi-signature of Claes-Jensen syndrome. This result served as a biomarker which independently highlighted KDM5C as the cause of the disorder in this patient. Because of the X-linked mode of inheritance, variant reclassification had a high impact on genetic counseling in this family. This example highlights the value of global methylome profiling in situations of variants of uncertain significance in genes with a known specific epi-signature.


Asunto(s)
Pérdida Auditiva Central , Discapacidad Intelectual , Atrofia Óptica , Metilación de ADN , Genes Ligados a X , Pérdida Auditiva Central/genética , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Humanos , Discapacidad Intelectual/genética , Masculino , Atrofia Óptica/genética
3.
Hum Mutat ; 43(12): 1837-1843, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35870179

RESUMEN

Synonymous variants have been shown to alter the correct splicing of pre-mRNAs and generate disease-causing transcripts. These variants are not an uncommon etiology of genetic disease; however, they are frequently overlooked during genetic testing in the absence of functional and clinical data. Here, we describe the occurrence of a synonymous variant [NM_005422.4 (TECTA):c.327C>T, p.(Gly109=)] in seven individuals with hearing loss from six unrelated families. The variant is not located near exonic/intronic boundaries but is predicted to impact splicing by activating a cryptic splicing donor site in exon 4 of TECTA. In vitro minigene assays show that the variant disrupts the reading frame of the canonical transcript, which is predicted to cause a premature termination codon 48 amino acids downstream of the variant, leading to nonsense-mediated decay. The variant is present in population databases, predominantly in Latinos of African ancestry, but is rare in other ethnic groups. Our findings suggest that this synonymous variant is likely pathogenic for TECTA-associated autosomal recessive hearing loss and seems to have arisen as a founder variant in this specific Latino subpopulation. This study demonstrates that synonymous variants need careful splicing assessment and support from additional testing methodologies to determine their clinical impact.


Asunto(s)
Sordera , Pérdida Auditiva , Humanos , Sitios de Empalme de ARN , Empalme del ARN/genética , Pérdida Auditiva/genética , Sordera/genética , Exones/genética , Proteínas de la Matriz Extracelular/genética , Proteínas Ligadas a GPI/genética
4.
Artículo en Inglés | MEDLINE | ID: mdl-35232817

RESUMEN

Li-Fraumeni syndrome (LFS) is one of the most common cancer predisposition syndromes that affects both children and adults. Individuals with LFS are at an increased risk of developing various types of cancer over their lifetime including soft tissue sarcomas, osteosarcomas, breast cancer, leukemia, brain tumors, and adrenocortical carcinoma. Heterozygous germline pathogenic variants in the tumor suppressor gene TP53 are the known causal genetic defect for LFS. Single-nucleotide variants (SNVs) including missense substitutions that occur in the highly conserved DNA binding domain of the protein are the most common alterations, followed by nonsense and splice site variants. Gross copy-number changes in TP53 are rare and account for <1% of all variants. Using next-generation sequencing (NGS) panels, we identified a paternally inherited germline intragenic duplication of TP53 in a child with metastatic osteosarcoma who later developed acute myeloid leukemia (AML). Transcriptome sequencing (RNA-seq) demonstrated the duplication was tandem, encompassing exons 2-6 and 28 nt of the untranslated region (UTR) upstream of the start codon in exon 2. The inclusion of the 28 nt is expected to result in a frameshift with a stop codon 18 codons downstream from the exon 6, leading to a loss-of-function allele. This case highlights the significance of simultaneous identification of both significant copy-number variants as well as SNVs/indels using NGS panels.


Asunto(s)
Neoplasias de la Corteza Suprarrenal , Neoplasias de la Mama , Síndrome de Li-Fraumeni , Proteína p53 Supresora de Tumor , Adulto , Neoplasias de la Mama/genética , Niño , Femenino , Duplicación de Gen/genética , Predisposición Genética a la Enfermedad , Mutación de Línea Germinal/genética , Humanos , Síndrome de Li-Fraumeni/genética , Proteína p53 Supresora de Tumor/genética
5.
Mol Genet Metab ; 135(1): 93-101, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34969639

RESUMEN

Mitochondrial disease diagnosis requires interrogation of both nuclear and mitochondrial (mtDNA) genomes for single-nucleotide variants (SNVs) and copy number alterations, both in the proband and often maternal relatives, together with careful phenotype correlation. We developed a comprehensive mtDNA sequencing test ('MitoGenome') using long-range PCR (LR-PCR) to amplify the full length of the mtDNA genome followed by next generation sequencing (NGS) to accurately detect SNVs and large-scale mtDNA deletions (LSMD), combined with droplet digital PCR (ddPCR) for LSMD heteroplasmy quantification. Overall, MitoGenome tests were performed on 428 samples from 394 patients with suspected or confirmed mitochondrial disease. The positive yield was 11% (43/394), including 34 patients with pathogenic or likely pathogenic SNVs (the most common being m.3243A > G in 8/34 (24%) patients), 8 patients with single LSMD, and 3 patients with multiple LSMD exceeding 10% heteroplasmy levels. Two patients with both LSMD and pathogenic SNV were detected. Overall, this LR-PCR/NGS assay provides a highly accurate and comprehensive diagnostic method for simultaneous mtDNA SNV detection at heteroplasmy levels as low as 1% and LSMD detection at heteroplasmy levels below 10%. Inclusion of maternal samples for variant classification and ddPCR to quantify LSMD heteroplasmy levels further enables accurate pathogenicity assessment and clinical correlation interpretation of mtDNA genome sequence variants and copy number alterations.


Asunto(s)
Genoma Mitocondrial , Enfermedades Mitocondriales , ADN Mitocondrial/genética , Genoma Mitocondrial/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mitocondrias/genética , Enfermedades Mitocondriales/diagnóstico , Enfermedades Mitocondriales/genética
6.
Cancer Genet ; 258-259: 110-119, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34710798

RESUMEN

The NTRK gene family is composed of NTRK1, NTRK2, and NTRK3, which encode three tropomyosin-receptor kinases, belonging to a class of tyrosine kinase receptors. These proteins are known to play roles in cell proliferation, differentiation, apoptosis, and survival. Fusions involving the NTRK genes are long known as drivers in many tumors. Although they occur in less than 5% of all malignancies, their occurrence in a great diversity of tumors has been documented. Several rare tumors including infantile fibrosarcoma, secretory breast carcinoma, and mammary analogue secretory carcinoma are accompanied by NTRK fusions in more than 90% of cases, demonstrating a diagnostic value for the NTRK fusion testing in these tumors. More recently, the development of effective targeted therapies has created a demand for their detection in all malignancies. A variety of approaches are available for testing including immunohistochemistry, fluorescence in situ hybridization (FISH), reverse transcription polymerase chain reaction (RT-PCR), and DNA- and RNA-based next-generation sequencing (NGS). This article reviews the molecular biology and tumorigenesis of NTRK fusions, their prevalence and clinical significance with a focus on available methods for fusion detection. The advantages and limitations of different technologies, the best practice algorithms for NTRK fusion detection, and the future direction of NTRK testing are also discussed.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias/diagnóstico , Proteínas de Fusión Oncogénica/genética , Receptor trkA/genética , Receptor trkB/genética , Receptor trkC/genética , Animales , Humanos , Neoplasias/genética , Pronóstico
8.
Invest Ophthalmol Vis Sci ; 62(3): 20, 2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33724292

RESUMEN

Purpose: Diabetic retinopathy (DR) remains a pressing issue worldwide. Abnormal angiogenesis is a distinct vascular lesion in DR, and research has established that vascular endothelial growth factor A (VEGF-A) is a primary mediator of such changes. However, limitations in current anti-VEGF therapies suggest that our understanding of molecular networks underlying ocular angiogenesis remains far from complete. Based on our long non-coding RNA (lncRNA) array analyses, HOX antisense intergenic RNA (HOTAIR) was identified as one of the top upregulated lncRNAs in high glucose-cultured human retinal endothelial cells (HRECs). Given the well-documented roles of HOTAIR in cancer, no studies have examined the epigenetic implications of HOTAIR in DR, and we investigated such relationships herein. Methods: We used HRECs exposed to various glucose concentrations and epigenetic modulators to examine HOTAIR, angiogenic, and DR-related molecular markers. Oxidative stress, angiogenesis, and mitochondrial dysfunction were assessed. Retinal tissues of diabetic rodents and the vitreous humor and serum of patients with proliferative DR were also investigated. Results: Hyperglycemia significantly augmented HOTAIR expression in HRECs and promoted angiogenesis, oxidative damage, and mitochondrial aberrations. Similarly, vitreous humor and serum from proliferative DR patients and retinas from diabetic animals demonstrated increased HOTAIR expression compared to non-diabetic controls. HOTAIR knockdown protected against glucose-induced increases of angiogenic and diabetes-associated molecules in the retina. Mechanistically, we showed that HOTAIR exerts its capabilities by preventing oxidative stress and modulating epigenetic pathways involving histone methylation, histone acetylation, DNA methylation, and transcription factors. Conclusions: Our findings suggest that HOTAIR is a critical lncRNA in the pathogenesis of DR and may potentially be important for diagnostic and therapeutic targeting.


Asunto(s)
Retinopatía Diabética/genética , Epigénesis Genética/genética , Regulación de la Expresión Génica/fisiología , ARN Largo no Codificante/genética , Neovascularización Retiniana/genética , Animales , Vasos Sanguíneos/fisiología , Células Cultivadas , Islas de CpG/genética , Metilación de ADN , Diabetes Mellitus Experimental , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Glucosa/farmacología , Humanos , Hibridación Fluorescente in Situ , Masculino , Ratones Endogámicos C57BL , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/metabolismo , Vasos Retinianos/patología
10.
Genet Med ; 23(6): 1065-1074, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33547396

RESUMEN

PURPOSE: We describe the clinical implementation of genome-wide DNA methylation analysis in rare disorders across the EpiSign diagnostic laboratory network and the assessment of results and clinical impact in the first subjects tested. METHODS: We outline the logistics and data flow between an integrated network of clinical diagnostics laboratories in Europe, the United States, and Canada. We describe the clinical validation of EpiSign using 211 specimens and assess the test performance and diagnostic yield in the first 207 subjects tested involving two patient subgroups: the targeted cohort (subjects with previous ambiguous/inconclusive genetic findings including genetic variants of unknown clinical significance) and the screening cohort (subjects with clinical findings consistent with hereditary neurodevelopmental syndromes and no previous conclusive genetic findings). RESULTS: Among the 207 subjects tested, 57 (27.6%) were positive for a diagnostic episignature including 48/136 (35.3%) in the targeted cohort and 8/71 (11.3%) in the screening cohort, with 4/207 (1.9%) remaining inconclusive after EpiSign analysis. CONCLUSION: This study describes the implementation of diagnostic clinical genomic DNA methylation testing in patients with rare disorders. It provides strong evidence of clinical utility of EpiSign analysis, including the ability to provide conclusive findings in the majority of subjects tested.


Asunto(s)
Metilación de ADN , Epigenómica , Canadá , Europa (Continente) , Humanos , Síndrome
11.
Int J Mol Sci ; 22(3)2021 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-33498634

RESUMEN

A growing number of genetic neurodevelopmental disorders are known to be associated with unique genomic DNA methylation patterns, called episignatures, which are detectable in peripheral blood. The intellectual developmental disorder, X-linked, syndromic, Armfield type (MRXSA) is caused by missense variants in FAM50A. Functional studies revealed the pathogenesis to be a spliceosomopathy that is characterized by atypical mRNA processing during development. In this study, we assessed the peripheral blood specimens in a cohort of individuals with MRXSA and detected a unique and highly specific DNA methylation episignature associated with this disorder. We used this episignature to construct a support vector machine model capable of sensitive and specific identification of individuals with pathogenic variants in FAM50A. This study contributes to the expanding number of genetic neurodevelopmental disorders with defined DNA methylation episignatures, provides an additional understanding of the associated molecular mechanisms, and further enhances our ability to diagnose patients with rare disorders.


Asunto(s)
Metilación de ADN , Discapacidad Intelectual Ligada al Cromosoma X/genética , Adulto , Estudios de Casos y Controles , Niño , Proteínas de Unión al ADN/genética , Epigenoma , Humanos , Masculino , Discapacidad Intelectual Ligada al Cromosoma X/etiología , Persona de Mediana Edad , Modelos Genéticos , Trastornos del Neurodesarrollo/genética , Proteínas de Unión al ARN/genética
12.
J Hum Genet ; 66(5): 451-464, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33093641

RESUMEN

The adaptation of a broad genomic sequencing approach in the clinical setting has been accompanied by considerations regarding the clinical utility, technical performance, and diagnostic yield compared to targeted genetic approaches. We have developed MedExome, an integrated framework for sequencing, variant calling (SNVs, Indels, and CNVs), and clinical assessment of ~4600 medically relevant genes. We compared the technical performance of MedExome with the whole-exome and targeted gene-panel sequencing, assessed the reasons for discordance, and evaluated the added clinical yield of MedExome in a cohort of unresolved subjects suspected of genetic disease. Our analysis showed that despite a higher average read depth in panels (3058 vs. 855), MedExome yielded full coverage of the enriched regions (>20X) and 99% variant concordance rate with panels. The discordance rate was associated with low-complexity regions, high-GC content, and low allele fractions, observed in both platforms. MedExome yielded full sensitivity in detecting clinically actionable variants, and the assessment of 138 patients with suspected genetic conditions resulted in 76 clinical reports (31 full [22.1%], 3 partial, and 42 uncertain/possible molecular diagnoses). MedExome sequencing has comparable performance in variant detection to gene panels. Added diagnostic yield justifies expanded implementation of broad genomic approaches in unresolved patients; however, cost-benefit and health systems impact warrants assessment.


Asunto(s)
Secuenciación del Exoma/métodos , Enfermedades Genéticas Congénitas/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Técnicas de Diagnóstico Molecular/métodos , Alelos , Composición de Base , Consanguinidad , Variaciones en el Número de Copia de ADN , Exoma , Biblioteca de Genes , Variación Genética , Homocigoto , Humanos , Mutación INDEL , Ontario , Mutación Puntual , Alineación de Secuencia , Flujo de Trabajo
13.
Int J Mol Sci ; 21(23)2020 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-33291301

RESUMEN

Mendelian neurodevelopmental disorders customarily present with complex and overlapping symptoms, complicating the clinical diagnosis. Individuals with a growing number of the so-called rare disorders exhibit unique, disorder-specific DNA methylation patterns, consequent to the underlying gene defects. Besides providing insights to the pathophysiology and molecular biology of these disorders, we can use these epigenetic patterns as functional biomarkers for the screening and diagnosis of these conditions. This review summarizes our current understanding of DNA methylation episignatures in rare disorders and describes the underlying technology and analytical approaches. We discuss the computational parameters, including statistical and machine learning methods, used for the screening and classification of genetic variants of uncertain clinical significance. Describing the rationale and principles applied to the specific computational models that are used to develop and adapt the DNA methylation episignatures for the diagnosis of rare disorders, we highlight the opportunities and challenges in this emerging branch of diagnostic medicine.


Asunto(s)
Metilación de ADN , Enfermedades Genéticas Congénitas/genética , Pruebas Genéticas/métodos , Trastornos del Neurodesarrollo/genética , Epigenoma , Enfermedades Genéticas Congénitas/diagnóstico , Pruebas Genéticas/normas , Humanos , Trastornos del Neurodesarrollo/diagnóstico
14.
Hum Mol Genet ; 29(R1): R27-R32, 2020 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-32644126

RESUMEN

The breadth and complexity of genetic testing in patients with suspected Mendelian neurodevelopmental disorders has rapidly expanded in the past two decades. However, in spite of advances in genomic technologies, genetic diagnosis remains elusive in more than half of these patients. Epigenomics, and in particular genomic DNA methylation profiles, are now known to be associated with the underpinning genetic defects in a growing number of Mendelian disorders. These often highly specific and sensitive molecular biomarkers have been used to screen these patient populations, resolve ambiguous clinical cases and interpret genetic variants of unknown clinical significance. Increasing the diagnostic yield beyond genomic sequencing technologies has rapidly propelled epigenomics to clinical utilization, with recent introduction of DNA methylation 'EpiSign' analysis in clinical diagnostic laboratories. This review provides an overview of the principles, applications and limitations of DNA methylation episignature analysis in patients with neurodevelopmental Mendelian disorders, and discusses clinical implications of this emerging diagnostic technology.


Asunto(s)
Biomarcadores/análisis , Metilación de ADN , Epigénesis Genética , Epigenómica , Variación Genética , Genoma , Trastornos del Neurodesarrollo/diagnóstico , Animales , Pruebas Genéticas , Humanos , Trastornos del Neurodesarrollo/genética , Fenotipo
15.
Genet Med ; 22(11): 1838-1850, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32694869

RESUMEN

PURPOSE: Nontruncating variants in SMARCA2, encoding a catalytic subunit of SWI/SNF chromatin remodeling complex, cause Nicolaides-Baraitser syndrome (NCBRS), a condition with intellectual disability and multiple congenital anomalies. Other disorders due to SMARCA2 are unknown. METHODS: By next-generation sequencing, we identified candidate variants in SMARCA2 in 20 individuals from 18 families with a syndromic neurodevelopmental disorder not consistent with NCBRS. To stratify variant interpretation, we functionally analyzed SMARCA2 variants in yeasts and performed transcriptomic and genome methylation analyses on blood leukocytes. RESULTS: Of 20 individuals, 14 showed a recognizable phenotype with recurrent features including epicanthal folds, blepharophimosis, and downturned nasal tip along with variable degree of intellectual disability (or blepharophimosis intellectual disability syndrome [BIS]). In contrast to most NCBRS variants, all SMARCA2 variants associated with BIS are localized outside the helicase domains. Yeast phenotype assays differentiated NCBRS from non-NCBRS SMARCA2 variants. Transcriptomic and DNA methylation signatures differentiated NCBRS from BIS and those with nonspecific phenotype. In the remaining six individuals with nonspecific dysmorphic features, clinical and molecular data did not permit variant reclassification. CONCLUSION: We identified a novel recognizable syndrome named BIS associated with clustered de novo SMARCA2 variants outside the helicase domains, phenotypically and molecularly distinct from NCBRS.


Asunto(s)
Blefarofimosis , Hipotricosis , Discapacidad Intelectual , Facies , Deformidades Congénitas del Pie , Humanos , Discapacidad Intelectual/genética , Fenotipo , Factores de Transcripción/genética
16.
J Hum Genet ; 65(10): 865-873, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32483276

RESUMEN

Poly-ADP-ribose-polymerase inhibitor (PARPi) treatment is indicated for advanced-stage ovarian tumors with BRCA1/2 deficiency. The "BRCAness" status is thought to be attributed to a tumor phenotype associated with a specific epigenomic DNA methylation profile. Here, we examined the diagnostic impact of combined BRCA1/2 sequence, copy number, and promoter DNA methylation analysis, and evaluated whether genomic DNA methylation patterns can predict the BRCAness in ovarian tumors. DNA sequencing of 172 human tissue samples of advanced-stage ovarian adenocarcinoma identified 36 samples with a clinically significant tier 1/2 sequence variants (point mutations and in/dels) and 9 samples with a CNV causing a loss of function in BRCA1/2. DNA methylation analysis of the promoter of BRCA1/2 identified promoter hypermethylation of BRCA1 in two mutation-negative samples. Computational modeling of genome-wide methylation markers, measured using Infinium EPIC arrays, resulted in a total accuracy of 0.75, sensitivity: 0.83, specificity: 0.64, positive predictive value: 0.76, negative predictive value: 0.74, and area under the receiver's operating curve (AUC): 0.77, in classifying tumors harboring a BRCA1/2 defect from the rest. These findings indicate that the assessment of CNV and promoter DNA methylation in BRCA1/2 increases the cumulative diagnostic yield by 10%, compared with the 20% yield achieved by sequence variant analysis alone. Genomic DNA methylation data can partially predict BRCAness in ovarian tumors; however, further investigation in expanded BRCA1/2 cohorts is needed, and the effect of other double strand DNA repair gene defects in these tumors warrants further investigations.


Asunto(s)
Adenocarcinoma/genética , Proteína BRCA1/genética , Proteína BRCA2/genética , Metilación de ADN , Genes BRCA1 , Genes BRCA2 , Técnicas de Diagnóstico Molecular , Síndromes Neoplásicos Hereditarios/genética , Neoplasias Ováricas/genética , Adenocarcinoma/diagnóstico , Adenocarcinoma/tratamiento farmacológico , Adulto , Anciano , Antineoplásicos/uso terapéutico , Área Bajo la Curva , Variaciones en el Número de Copia de ADN , ADN de Neoplasias/química , ADN de Neoplasias/genética , Femenino , Predisposición Genética a la Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Mutación INDEL , Persona de Mediana Edad , Síndromes Neoplásicos Hereditarios/diagnóstico , Síndromes Neoplásicos Hereditarios/tratamiento farmacológico , Neoplasias Ováricas/diagnóstico , Neoplasias Ováricas/tratamiento farmacológico , Mutación Puntual , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Valor Predictivo de las Pruebas , Regiones Promotoras Genéticas/genética , Curva ROC , Sensibilidad y Especificidad
17.
Am J Physiol Cell Physiol ; 319(2): C268-C276, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32459505

RESUMEN

DNA methylation, a critical epigenetic mechanism, plays an important role in governing gene expressions during biological processes such as aging, which is well known to be accelerated in hyperglycemia (diabetes). In the present study, we investigated the effects of glucose on whole genome DNA methylation in small [human retinal microvascular endothelial cells (HRECs)] and large [human umbilical vein endothelial cells (HUVECs)] vessel endothelial cell (EC) lines exposed to basal or high glucose-containing media for variable lengths of time. Using the Infinium EPIC array, we obtained 773,133 CpG sites (probes) for analysis. Unsupervised clustering of the top 5% probes identified four distinct clusters within EC groups, with significant methylation differences attributed to EC types and the duration of cell culture rather than glucose stimuli alone. When comparing the ECs incubated for 2 days versus 7 days, hierarchical clustering analyses [methylation change >10% and false discovery rate (FDR) <0.05] identified 17,354 and 128 differentially methylated CpGs for HUVECs and HRECs, respectively. Predominant DNA hypermethylation was associated with the length of culture and was enriched for gene enhancer elements and regions surrounding CpG shores and shelves. We identified 88 differentially methylated regions (DMRs) for HUVECs and 8 DMRs for HRECs (all FDR <0.05). Pathway enrichment analyses of DMRs highlighted involvement of regulators of embryonic development (i.e., HOX genes) and cellular differentiation [transforming growth factor-ß (TGF-ß) family members]. Collectively, our findings suggest that DNA methylation is a complex process that involves tightly coordinated, cell-specific mechanisms. Such changes in methylation overlap genes critical for cellular differentiation and embryonic development.


Asunto(s)
Envejecimiento/genética , Islas de CpG/genética , Metilación de ADN/genética , Células Endoteliales/metabolismo , Envejecimiento/patología , Islas de CpG/efectos de los fármacos , ADN/genética , Metilación de ADN/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Epigénesis Genética , Regulación de la Expresión Génica/efectos de los fármacos , Genes Homeobox/genética , Genoma Humano/genética , Glucosa/farmacología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Regiones Promotoras Genéticas/efectos de los fármacos
18.
Am J Hum Genet ; 106(3): 356-370, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32109418

RESUMEN

Genetic syndromes frequently present with overlapping clinical features and inconclusive or ambiguous genetic findings which can confound accurate diagnosis and clinical management. An expanding number of genetic syndromes have been shown to have unique genomic DNA methylation patterns (called "episignatures"). Peripheral blood episignatures can be used for diagnostic testing as well as for the interpretation of ambiguous genetic test results. We present here an approach to episignature mapping in 42 genetic syndromes, which has allowed the identification of 34 robust disease-specific episignatures. We examine emerging patterns of overlap, as well as similarities and hierarchical relationships across these episignatures, to highlight their key features as they are related to genetic heterogeneity, dosage effect, unaffected carrier status, and incomplete penetrance. We demonstrate the necessity of multiclass modeling for accurate genetic variant classification and show how disease classification using a single episignature at a time can sometimes lead to classification errors in closely related episignatures. We demonstrate the utility of this tool in resolving ambiguous clinical cases and identification of previously undiagnosed cases through mass screening of a large cohort of subjects with developmental delays and congenital anomalies. This study more than doubles the number of published syndromes with DNA methylation episignatures and, most significantly, opens new avenues for accurate diagnosis and clinical assessment in individuals affected by these disorders.


Asunto(s)
Metilación de ADN , Trastornos del Neurodesarrollo/genética , Fenotipo , Estudios de Cohortes , Heterogeneidad Genética , Humanos , Síndrome
19.
Clin Epigenetics ; 12(1): 7, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31910894

RESUMEN

BACKGROUND: We previously associated HIST1H1E mutations causing Rahman syndrome with a specific genome-wide methylation pattern. RESULTS: Methylome analysis from peripheral blood samples of six affected subjects led us to identify a specific hypomethylated profile. This "episignature" was enriched for genes involved in neuronal system development and function. A computational classifier yielded full sensitivity and specificity in detecting subjects with Rahman syndrome. Applying this model to a cohort of undiagnosed probands allowed us to reach diagnosis in one subject. CONCLUSIONS: We demonstrate an epigenetic signature in subjects with Rahman syndrome that can be used to reach molecular diagnosis.


Asunto(s)
Metilación de ADN , Discapacidades del Desarrollo/genética , Mutación del Sistema de Lectura , Histonas/genética , Discapacidad Intelectual/genética , Encéfalo/metabolismo , Discapacidades del Desarrollo/metabolismo , Epigénesis Genética , Humanos , Discapacidad Intelectual/metabolismo , Neuronas/metabolismo , Transducción de Señal/genética , Síndrome
20.
Genome Biol ; 20(1): 146, 2019 08 14.
Artículo en Inglés | MEDLINE | ID: mdl-31409373

RESUMEN

BACKGROUND: Epigenetic clocks are mathematical models that predict the biological age of an individual using DNA methylation data and have emerged in the last few years as the most accurate biomarkers of the aging process. However, little is known about the molecular mechanisms that control the rate of such clocks. Here, we have examined the human epigenetic clock in patients with a variety of developmental disorders, harboring mutations in proteins of the epigenetic machinery. RESULTS: Using the Horvath epigenetic clock, we perform an unbiased screen for epigenetic age acceleration in the blood of these patients. We demonstrate that loss-of-function mutations in the H3K36 histone methyltransferase NSD1, which cause Sotos syndrome, substantially accelerate epigenetic aging. Furthermore, we show that the normal aging process and Sotos syndrome share methylation changes and the genomic context in which they occur. Finally, we found that the Horvath clock CpG sites are characterized by a higher Shannon methylation entropy when compared with the rest of the genome, which is dramatically decreased in Sotos syndrome patients. CONCLUSIONS: These results suggest that the H3K36 methylation machinery is a key component of the epigenetic maintenance system in humans, which controls the rate of epigenetic aging, and this role seems to be conserved in model organisms. Our observations provide novel insights into the mechanisms behind the epigenetic aging clock and we expect will shed light on the different processes that erode the human epigenetic landscape during aging.


Asunto(s)
Envejecimiento/genética , Relojes Biológicos/genética , Epigénesis Genética , Pruebas Genéticas , N-Metiltransferasa de Histona-Lisina/genética , Histonas/metabolismo , Lisina/metabolismo , Adulto , Islas de CpG/genética , Metilación de ADN/genética , Entropía , Genoma Humano , Humanos , Lactante , Modelos Genéticos , Síndrome de Sotos/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...