Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
J Mol Diagn ; 25(3): 156-167, 2023 03.
Article in English | MEDLINE | ID: mdl-36563937

ABSTRACT

Nearly 14% of disease-causing germline variants result from the disruption of mRNA splicing. Most (67%) DNA variants predicted in silico to disrupt splicing are classified as variants of uncertain significance. An analytic workflow-splice effect event resolver (SPEER)-was developed and validated to use mRNA sequencing to reveal significant deviations in splicing, pinpoint the DNA variants potentially involved, and measure the deleterious effects of the altered splicing on mRNA transcripts, providing evidence for assessing the pathogenicity of the variant. SPEER was used to analyze leukocyte RNA encoding 63 hereditary cancer syndrome-related genes in 20,317 patients. Among 3563 patients (17.5%) with at least one DNA variant predicted to affect splicing, 971 (4.8%) had altered splicing with a deleterious effect on the transcript, and 40 had altered splicing due to a DNA variant located outside of the reportable range of the test. Integrating SPEER results into the interpretation of variants allowed variants of uncertain significance to be reclassified as pathogenic or likely pathogenic in 0.4%, and as benign or likely benign in 5.9%, of the 20,317 patients. SPEER-based evidence was associated with a significantly greater effect on classifications of pathogenic or likely pathogenic and benign or likely benign in nonwhite versus non-Hispanic white patients, illustrating that evidence derived from mRNA splicing analysis may help to reduce ethnic/ancestral disparities in genetic testing.


Subject(s)
Genetic Testing , Neoplastic Syndromes, Hereditary , Humans , Genetic Testing/methods , RNA Splicing , RNA, Messenger/genetics , RNA , Neoplastic Syndromes, Hereditary/genetics
2.
Clin Cancer Res ; 24(15): 3539-3549, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29691297

ABSTRACT

Purpose: To analytically and clinically validate a circulating cell-free tumor DNA sequencing test for comprehensive tumor genotyping and demonstrate its clinical feasibility.Experimental Design: Analytic validation was conducted according to established principles and guidelines. Blood-to-blood clinical validation comprised blinded external comparison with clinical droplet digital PCR across 222 consecutive biomarker-positive clinical samples. Blood-to-tissue clinical validation comprised comparison of digital sequencing calls to those documented in the medical record of 543 consecutive lung cancer patients. Clinical experience was reported from 10,593 consecutive clinical samples.Results: Digital sequencing technology enabled variant detection down to 0.02% to 0.04% allelic fraction/2.12 copies with ≤0.3%/2.24-2.76 copies 95% limits of detection while maintaining high specificity [prevalence-adjusted positive predictive values (PPV) >98%]. Clinical validation using orthogonal plasma- and tissue-based clinical genotyping across >750 patients demonstrated high accuracy and specificity [positive percent agreement (PPAs) and negative percent agreement (NPAs) >99% and PPVs 92%-100%]. Clinical use in 10,593 advanced adult solid tumor patients demonstrated high feasibility (>99.6% technical success rate) and clinical sensitivity (85.9%), with high potential actionability (16.7% with FDA-approved on-label treatment options; 72.0% with treatment or trial recommendations), particularly in non-small cell lung cancer, where 34.5% of patient samples comprised a directly targetable standard-of-care biomarker.Conclusions: High concordance with orthogonal clinical plasma- and tissue-based genotyping methods supports the clinical accuracy of digital sequencing across all four types of targetable genomic alterations. Digital sequencing's clinical applicability is further supported by high rates of technical success and biomarker target discovery. Clin Cancer Res; 24(15); 3539-49. ©2018 AACR.


Subject(s)
Cell-Free Nucleic Acids/genetics , Circulating Tumor DNA/genetics , Genomics , Neoplasms/genetics , Biomarkers, Tumor , Cell-Free Nucleic Acids/blood , Circulating Tumor DNA/blood , Female , Genotype , Genotyping Techniques , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation , Neoplasms/blood , Neoplasms/pathology
3.
BMC Bioinformatics ; 8: 274, 2007 Jul 30.
Article in English | MEDLINE | ID: mdl-17663765

ABSTRACT

BACKGROUND: Short (~5 nucleotides) interspersed repeats regulate several aspects of post-transcriptional gene expression. Previously we developed an algorithm (REPFIND) that assigns P-values to all repeated motifs in a given nucleic acid sequence and reliably identifies clusters of short CAC-containing motifs required for mRNA localization in Xenopus oocytes. DESCRIPTION: In order to facilitate the identification of genes possessing clusters of repeats that regulate post-transcriptional aspects of gene expression in mammalian genes, we used REPFIND to create a database of all repeated motifs in the 3' untranslated regions (UTR) of genes from the Mammalian Gene Collection (MGC). The MGC database includes seven vertebrate species: human, cow, rat, mouse and three non-mammalian vertebrate species. A web-based application was developed to search this database of repeated motifs to generate species-specific lists of genes containing specific classes of repeats in their 3'-UTRs. This computational tool is called 3'-UTR SIRF (Short Interspersed Repeat Finder), and it reveals that hundreds of human genes contain an abundance of short CAC-rich and CAG-rich repeats in their 3'-UTRs that are similar to those found in mRNAs localized to the neurites of neurons. We tested four candidate mRNAs for localization in rat hippocampal neurons by in situ hybridization. Our results show that two candidate CAC-rich (Syntaxin 1B and Tubulin beta4) and two candidate CAG-rich (Sec61alpha and Syntaxin 1A) mRNAs are localized to distal neurites, whereas two control mRNAs lacking repeated motifs in their 3'-UTR remain primarily in the cell body. CONCLUSION: Computational data generated with 3'-UTR SIRF indicate that hundreds of mammalian genes have an abundance of short CA-containing motifs that may direct mRNA localization in neurons. In situ hybridization shows that four candidate mRNAs are localized to distal neurites of cultured hippocampal neurons. These data suggest that short CA-containing motifs may be part of a widely utilized genetic code that regulates mRNA localization in vertebrate cells. The use of 3'-UTR SIRF to search for new classes of motifs that regulate other aspects of gene expression should yield important information in future studies addressing cis-regulatory information located in 3'-UTRs.


Subject(s)
3' Untranslated Regions/genetics , Databases, Genetic , Interspersed Repetitive Sequences/genetics , Multigene Family/genetics , Sequence Analysis, DNA/methods , Base Sequence , Molecular Sequence Data
4.
Stem Cells Dev ; 24(13): 1536-45, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25749005

ABSTRACT

Removal of cytosine methylation from the genome is critical for reprogramming and transdifferentiation and plays a central role in our understanding of the fundamental principles of embryo lineage development. One of the major models for studying cytosine demethylation is the mammalian germ line during the primordial germ cell (PGC) stage of embryo development. It is now understood that oxidation of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) is required to remove cytosine methylation in a locus-specific manner in PGCs; however, the mechanisms downstream of 5hmC are controversial and hypothesized to involve either active demethylation or replication-coupled loss. In the current study, we used the aorta-gonad-mesonephros (AGM) organ culture model to show that this model recapitulates germ line reprogramming, including 5hmC reorganization and loss of cytosine methylation from Snrpn and H19 imprinting control centers (ICCs). To directly address the hypothesis that cell proliferation is required for cytosine demethylation, we blocked PI3-kinase-dependent PGC proliferation and show that this leads to a G1 and G2/M cell cycle arrest in PGCs, together with retained levels of cytosine methylation at the Snrpn ICC, but not at the H19 ICC. Taken together, the AGM organ culture model is an important tool to evaluate mechanisms of locus-specific demethylation and the role of PI3-kinase-dependent PGC proliferation in the locus-specific removal of cytosine methylation from the genome.


Subject(s)
Aorta/metabolism , DNA Methylation , DNA Replication , Germ Cells/metabolism , Gonads/metabolism , Mesonephros/metabolism , 5-Methylcytosine/metabolism , Animals , Aorta/cytology , Cell Proliferation , Germ Cells/physiology , Gonads/cytology , Mesonephros/cytology , Mice , Organ Culture Techniques
5.
Nat Cell Biol ; 15(1): 113-22, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23242216

ABSTRACT

The generation of research-quality, clinically relevant cell types in vitro from human pluripotent stem cells requires a detailed understanding of the equivalent human cell types. Here we analysed 134 human embryonic and fetal samples from 6 to 20 developmental weeks and identified the stages at which cKIT(+) primordial germ cells (PGCs), the precursors of gametes, undergo whole-genome epigenetic reprogramming with global depletion of 5mC, H3K27me3 and H2A.Z, and the time at which imprint erasure is initiated and 5hmC is present. Using five alternative in vitro differentiation strategies combined with single-cell microfluidic analysis and a bona fide human cKIT(+) PGC signature, we show the stage of cKIT(+) PGC formation in the first 16 days of differentiation. Taken together, our study creates a resource of human germ line ontogeny that is essential for future studies aimed at in vitro differentiation and unveiling the mechanisms necessary to pass human DNA from one generation to the next.


Subject(s)
Cell Differentiation , Genomic Imprinting , Germ Cells/metabolism , Germ Cells/physiology , Proto-Oncogene Proteins c-kit/metabolism , Cells, Cultured , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA Methylation , Embryo, Mammalian/cytology , Embryonic Stem Cells/metabolism , Female , Fetus/cytology , Gene Expression Regulation, Developmental , Histones/metabolism , Humans , Male , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Ovary/cytology , Ovary/embryology , Ovary/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-kit/genetics , Single-Cell Analysis , Testis/cytology , Testis/embryology , Testis/metabolism , Transcriptome
6.
Cell Stem Cell ; 12(4): 470-8, 2013 Apr 04.
Article in English | MEDLINE | ID: mdl-23415914

ABSTRACT

Primordial germ cells (PGCs) undergo dramatic rearrangements to their methylome during embryogenesis, including initial genome-wide DNA demethylation that establishes the germline epigenetic ground state. The role of the 5-methylcytosine (5mC) dioxygenases Tet1 and Tet2 in the initial genome-wide DNA demethylation process has not been examined directly. Using PGCs differentiated from either control or Tet2(-/-); Tet1 knockdown embryonic stem cells (ESCs), we show that in vitro PGC (iPGC) formation and genome-wide DNA demethylation are unaffected by the absence of Tet1 and Tet2, and thus 5-hydroxymethylcytosine (5hmC). However, numerous promoters and gene bodies were hypermethylated in mutant iPGCs, which is consistent with a role for 5hmC as an intermediate in locus-specific demethylation. Altogether, our results support a revised model of PGC DNA demethylation in which the first phase of comprehensive 5mC loss does not involve 5hmC. Instead, Tet1 and Tet2 have a locus-specific role in shaping the PGC epigenome during subsequent development.


Subject(s)
DNA Methylation , DNA-Binding Proteins/metabolism , Germ Cells/metabolism , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation , Cytosine/analogs & derivatives , Cytosine/metabolism , Dioxygenases , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Genome/genetics , Germ Cells/cytology , Green Fluorescent Proteins/metabolism , Mice
7.
PLoS One ; 6(1): e16478, 2011 Jan 27.
Article in English | MEDLINE | ID: mdl-21304588

ABSTRACT

Pluripotent stem cells (PSCs) hold significant promise in regenerative medicine due to their unlimited capacity for self-renewal and potential to differentiate into every cell type in the body. One major barrier to the use of PSCs is their potential risk for tumor initiation following differentiation and transplantation in vivo. In the current study we sought to evaluate the role of the tumor suppressor Pten in murine PSC neoplastic progression. Using eight functional assays that have previously been used to indicate PSC adaptation or transformation, Pten null embryonic stem cells (ESCs) failed to rate as significant in five of them. Instead, our data demonstrate that the loss of Pten causes the emergence of a small number of aggressive, teratoma-initiating embryonic carcinoma cells (ECCs) during differentiation in vitro, while the remaining 90-95% of differentiated cells are non-tumorigenic. Furthermore, our data show that the mechanism by which Pten null ECCs emerge in vitro and cause tumors in vivo is through increased survival and self-renewal, due to failed repression of the transcription factor Nanog.


Subject(s)
Cell Differentiation , Cell Transformation, Neoplastic , Homeodomain Proteins/physiology , PTEN Phosphohydrolase/deficiency , Pluripotent Stem Cells/pathology , Animals , Cell Survival , Embryonic Stem Cells , Mice , Nanog Homeobox Protein , Neoplastic Stem Cells/pathology
8.
PLoS One ; 6(12): e28960, 2011.
Article in English | MEDLINE | ID: mdl-22194959

ABSTRACT

The cell intrinsic programming that regulates mammalian primordial germ cell (PGC) development in the pre-gonadal stage is challenging to investigate. To overcome this we created a transgene-free method for generating PGCs in vitro (iPGCs) from mouse embryonic stem cells (ESCs). Using labeling for SSEA1 and cKit, two cell surface molecules used previously to isolate presumptive iPGCs, we show that not all SSEA1+/cKit+ double positive cells exhibit a PGC identity. Instead, we determined that selecting for cKit(bright) cells within the SSEA1+ fraction significantly enriches for the putative iPGC population. Single cell analysis comparing SSEA1+/cKit(bright) iPGCs to ESCs and embryonic PGCs demonstrates that 97% of single iPGCs co-express PGC signature genes Blimp1, Stella, Dnd1, Prdm14 and Dazl at similar levels to e9.5-10.5 PGCs, whereas 90% of single mouse ESC do not co-express PGC signature genes. For the 10% of ESCs that co-express PGC signature genes, the levels are significantly lower than iPGCs. Microarray analysis shows that iPGCs are transcriptionally distinct from ESCs and repress gene ontology groups associated with mesoderm and heart development. At the level of chromatin, iPGCs contain 5-methyl cytosine bases in their DNA at imprinted and non-imprinted loci, and are enriched in histone H3 lysine 27 trimethylation, yet do not have detectable levels of Mvh protein, consistent with a Blimp1-positive pre-gonadal PGC identity. In order to determine whether iPGC formation is dependent upon Blimp1, we generated Blimp1 null ESCs and found that loss of Blimp1 significantly depletes SSEA1/cKit(bright) iPGCs. Taken together, the generation of Blimp1-positive iPGCs from ESCs constitutes a robust model for examining cell-intrinsic regulation of PGCs during the Blimp1-positive stage of development.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Germ Cells/cytology , Germ Cells/metabolism , Single-Cell Analysis/methods , Transcription Factors/metabolism , Animals , Biomarkers/metabolism , Cell Differentiation/genetics , Cell Separation , Embryoid Bodies/cytology , Embryoid Bodies/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gonads/cytology , Lewis X Antigen/metabolism , Mesoderm/metabolism , Mice , Models, Biological , Octamer Transcription Factor-3/metabolism , Oligonucleotide Array Sequence Analysis , Positive Regulatory Domain I-Binding Factor 1 , Proto-Oncogene Proteins c-kit/metabolism , Transcription, Genetic , Transgenes
SELECTION OF CITATIONS
SEARCH DETAIL