Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
2.
NAR Genom Bioinform ; 4(4): lqac075, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36267125

ABSTRACT

Score-based motif enrichment analysis (MEA) is typically applied to regulatory DNA to infer transcription factors (TFs) that may modulate transcription and chromatin state in different conditions. Most MEA methods determine motif enrichment independent of motif position within a sequence, even when those sequences harbor anchor points that motifs and their bound TFs may functionally interact with in a distance-dependent fashion, such as other TF binding motifs, transcription start sites (TSS), sequencing assay cleavage sites, or other biologically meaningful features. We developed motif enrichment positional profiling (MEPP), a novel MEA method that outputs a positional enrichment profile of a given TF's binding motif relative to key anchor points (e.g. transcription start sites, or other motifs) within the analyzed sequences while accounting for lower-order nucleotide bias. Using transcription initiation and TF binding as test cases, we demonstrate MEPP's utility in determining the sequence positions where motif presence correlates with measures of biological activity, inferring positional dependencies of binding site function. We demonstrate how MEPP can be applied to interpretation and hypothesis generation from experiments that quantify transcription initiation, chromatin structure, or TF binding measurements. MEPP is available for download from https://github.com/npdeloss/mepp.

3.
BMC Bioinformatics ; 21(1): 410, 2020 Sep 16.
Article in English | MEDLINE | ID: mdl-32938397

ABSTRACT

BACKGROUND: Motif enrichment analysis (MEA) identifies over-represented transcription factor binding (TF) motifs in the DNA sequence of regulatory regions, enabling researchers to infer which transcription factors can regulate transcriptional response to a stimulus, or identify sequence features found near a target protein in a ChIP-seq experiment. Score-based MEA determines motifs enriched in regions exhibiting extreme differences in regulatory activity, but existing methods do not control for biases in GC content or dinucleotide composition. This lack of control for sequence bias, such as those often found in CpG islands, can obscure the enrichment of biologically relevant motifs. RESULTS: We developed Motif Enrichment In Ranked Lists of Peaks (MEIRLOP), a novel MEA method that determines enrichment of TF binding motifs in a list of scored regulatory regions, while controlling for sequence bias. In this study, we compare MEIRLOP against other MEA methods in identifying binding motifs found enriched in differentially active regulatory regions after interferon-beta stimulus, finding that using logistic regression and covariates improves the ability to call enrichment of ISGF3 binding motifs from differential acetylation ChIP-seq data compared to other methods. Our method achieves similar or better performance compared to other methods when quantifying the enrichment of TF binding motifs from ENCODE TF ChIP-seq datasets. We also demonstrate how MEIRLOP is broadly applicable to the analysis of numerous types of NGS assays and experimental designs. CONCLUSIONS: Our results demonstrate the importance of controlling for sequence bias when accurately identifying enriched DNA sequence motifs using score-based MEA. MEIRLOP is available for download from https://github.com/npdeloss/meirlop under the MIT license.


Subject(s)
Computational Biology/methods , Nucleotide Motifs/genetics , Sequence Analysis, DNA/methods , Bias , Humans
4.
Cell ; 179(2): 373-391.e27, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31585079

ABSTRACT

Cells regulate gene expression in response to salient external stimuli. In neurons, depolarization leads to the expression of inducible transcription factors (ITFs) that direct subsequent gene regulation. Depolarization encodes both a neuron's action potential (AP) output and synaptic inputs, via excitatory postsynaptic potentials (EPSPs). However, it is unclear if distinct types of electrical activity can be transformed by an ITF into distinct modes of genomic regulation. Here, we show that APs and EPSPs in mouse hippocampal neurons trigger two spatially segregated and molecularly distinct induction mechanisms that lead to the expression of the ITF NPAS4. These two pathways culminate in the formation of stimulus-specific NPAS4 heterodimers that exhibit distinct DNA binding patterns. Thus, NPAS4 differentially communicates increases in a neuron's spiking output and synaptic inputs to the nucleus, enabling gene regulation to be tailored to the type of depolarizing activity along the somato-dendritic axis of a neuron.


Subject(s)
Action Potentials , Basic Helix-Loop-Helix Transcription Factors/genetics , Excitatory Postsynaptic Potentials , Neurons/metabolism , Transcriptional Activation , 3' Untranslated Regions , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , CA1 Region, Hippocampal/cytology , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/physiology , Cells, Cultured , Female , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Protein Multimerization , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley
5.
J Mol Diagn ; 21(3): 437-448, 2019 05.
Article in English | MEDLINE | ID: mdl-30731207

ABSTRACT

Inherited cardiomyopathies (ICs) are a major cause of heart disease. Given their marked clinical and genetic heterogeneity, the content and clinical utility of IC multi-gene panels has been the topic of continuous debate. Our genetics diagnostic laboratory has been providing clinical diagnostic testing for ICs since 2012. We began by testing nine genes and expanded our panel by fivefold in 2015. Here, we describe the implementation of a cost-effective next-generation sequencing (NGS)-based assay for testing of IC genes, including a protocol that minimizes the amount of Sanger sequencing required to confirm variants identified by NGS, which reduces the cost and time of testing. The NGS assay was developed for the simultaneous analysis of 45 IC genes and was assessed for the impact of panel expansion on variant detection, turnaround time, and cost of testing in a cohort of 993 patients. The assay led to a considerable reduction in test cost and turnaround time. However, only a marginal increase was observed in the diagnostic yield, whereas the rate of inconclusive findings increased considerably. These findings suggest that the ongoing evaluation of gene content and monitoring of clinical utility for multi-gene tests are essential to achieve maximum clinical utility of multi-gene tests in a publicly funded health care setting.


Subject(s)
Cardiomyopathies/diagnosis , Cardiomyopathies/genetics , Delivery of Health Care , Genetic Testing , Inheritance Patterns/genetics , Molecular Diagnostic Techniques , High-Throughput Nucleotide Sequencing/standards , Humans , Reproducibility of Results , Sensitivity and Specificity , Sequence Analysis, DNA/standards
6.
Nat Commun ; 8(1): 1922, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29203771

ABSTRACT

Despite novel therapies, relapse of multiple myeloma (MM) is virtually inevitable. Amplification of chromosome 1q, which harbors the inflammation-responsive RNA editase adenosine deaminase acting on RNA (ADAR)1 gene, occurs in 30-50% of MM patients and portends a poor prognosis. Since adenosine-to-inosine RNA editing has recently emerged as a driver of cancer progression, genomic amplification combined with inflammatory cytokine activation of ADAR1 could stimulate MM progression and therapeutic resistance. Here, we report that high ADAR1 RNA expression correlates with reduced patient survival rates in the MMRF CoMMpass data set. Expression of wild-type, but not mutant, ADAR1 enhances Alu-dependent editing and transcriptional activity of GLI1, a Hedgehog (Hh) pathway transcriptional activator and self-renewal agonist, and promotes immunomodulatory drug resistance in vitro. Finally, ADAR1 knockdown reduces regeneration of high-risk MM in serially transplantable patient-derived xenografts. These data demonstrate that ADAR1 promotes malignant regeneration of MM and if selectively inhibited may obviate progression and relapse.


Subject(s)
Adenosine Deaminase/genetics , Multiple Myeloma/genetics , Neoplasm Recurrence, Local/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Zinc Finger Protein GLI1/metabolism , Adenosine Deaminase/metabolism , Adult , Aged , Animals , Case-Control Studies , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Gene Knockdown Techniques , Humans , In Vitro Techniques , Male , Mice , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neoplasm Recurrence, Local/metabolism , Neoplasm Transplantation , Prognosis , RNA Editing/genetics , RNA-Binding Proteins/metabolism
7.
Cell Stem Cell ; 19(5): 599-612, 2016 11 03.
Article in English | MEDLINE | ID: mdl-27570067

ABSTRACT

Age-related human hematopoietic stem cell (HSC) exhaustion and myeloid-lineage skewing promote oncogenic transformation of hematopoietic progenitor cells into therapy-resistant leukemia stem cells (LSCs) in secondary acute myeloid leukemia (AML). While acquisition of clonal DNA mutations has been linked to increased rates of secondary AML for individuals older than 60 years, the contribution of RNA processing alterations to human hematopoietic stem and progenitor aging and LSC generation remains unclear. Comprehensive RNA sequencing and splice-isoform-specific PCR uncovered characteristic RNA splice isoform expression patterns that distinguished normal young and aged human stem and progenitor cells (HSPCs) from malignant myelodysplastic syndrome (MDS) and AML progenitors. In splicing reporter assays and pre-clinical patient-derived AML models, treatment with a pharmacologic splicing modulator, 17S-FD-895, reversed pro-survival splice isoform switching and significantly impaired LSC maintenance. Therapeutic splicing modulation, together with monitoring splice isoform biomarkers of healthy HSPC aging versus LSC generation, may be employed safely and effectively to prevent relapse, the leading cause of leukemia-related mortality.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA Splicing/genetics , Animals , Cell Survival/genetics , Cellular Senescence/genetics , Coculture Techniques , HEK293 Cells , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Mice , Myelodysplastic Syndromes/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Spliceosomes/metabolism , Stromal Cells/metabolism
8.
Cell Stem Cell ; 19(2): 177-191, 2016 08 04.
Article in English | MEDLINE | ID: mdl-27292188

ABSTRACT

Post-transcriptional adenosine-to-inosine RNA editing mediated by adenosine deaminase acting on RNA1 (ADAR1) promotes cancer progression and therapeutic resistance. However, ADAR1 editase-dependent mechanisms governing leukemia stem cell (LSC) generation have not been elucidated. In blast crisis chronic myeloid leukemia (BC CML), we show that increased JAK2 signaling and BCR-ABL1 amplification activate ADAR1. In a humanized BC CML mouse model, combined JAK2 and BCR-ABL1 inhibition prevents LSC self-renewal commensurate with ADAR1 downregulation. Lentiviral ADAR1 wild-type, but not an editing-defective ADAR1(E912A) mutant, induces self-renewal gene expression and impairs biogenesis of stem cell regulatory let-7 microRNAs. Combined RNA sequencing, qRT-PCR, CLIP-ADAR1, and pri-let-7 mutagenesis data suggest that ADAR1 promotes LSC generation via let-7 pri-microRNA editing and LIN28B upregulation. A small-molecule tool compound antagonizes ADAR1's effect on LSC self-renewal in stromal co-cultures and restores let-7 biogenesis. Thus, ADAR1 activation represents a unique therapeutic vulnerability in LSCs with active JAK2 signaling.


Subject(s)
Adenosine Deaminase/metabolism , Cell Self Renewal , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , MicroRNAs/metabolism , RNA-Binding Proteins/metabolism , Adenosine Deaminase/genetics , Animals , Base Sequence , Cell Self Renewal/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Janus Kinase 2/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Mice , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , RNA Editing/genetics , RNA-Binding Proteins/genetics , Signal Transduction/genetics
9.
Sci Rep ; 6: 23885, 2016 Apr 04.
Article in English | MEDLINE | ID: mdl-27041210

ABSTRACT

While implicated in therapeutic resistance, malignant progenitor cell cycle kinetics have been difficult to quantify in real-time. We developed an efficient lentiviral bicistronic fluorescent, ubiquitination-based cell cycle indicator reporter (Fucci2BL) to image live single progenitors on a defined niche coupled with cell cycle gene expression analysis. We have identified key differences in cell cycle regulatory gene expression and transit times between normal and chronic myeloid leukemia progenitors that may inform cancer stem cell eradication strategies.


Subject(s)
Gene Expression Profiling/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnostic imaging , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Neoplastic Stem Cells/metabolism , Sequence Analysis, RNA/methods , Stem Cell Niche , Cell Cycle , Cell Line , Coculture Techniques , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Humans , Kinetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Microscopy, Confocal , Time-Lapse Imaging
10.
Proc Natl Acad Sci U S A ; 112(50): 15444-9, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26621726

ABSTRACT

Formative research suggests that a human embryonic stem cell-specific alternative splicing gene regulatory network, which is repressed by Muscleblind-like (MBNL) RNA binding proteins, is involved in cell reprogramming. In this study, RNA sequencing, splice isoform-specific quantitative RT-PCR, lentiviral transduction, and in vivo humanized mouse model studies demonstrated that malignant reprogramming of progenitors into self-renewing blast crisis chronic myeloid leukemia stem cells (BC LSCs) was partially driven by decreased MBNL3. Lentiviral knockdown of MBNL3 resulted in reversion to an embryonic alternative splice isoform program typified by overexpression of CD44 transcript variant 3, containing variant exons 8-10, and BC LSC proliferation. Although isoform-specific lentiviral CD44v3 overexpression enhanced chronic phase chronic myeloid leukemia (CML) progenitor replating capacity, lentiviral shRNA knockdown abrogated these effects. Combined treatment with a humanized pan-CD44 monoclonal antibody and a breakpoint cluster region - ABL proto-oncogene 1, nonreceptor tyrosine kinase (BCR-ABL1) antagonist inhibited LSC maintenance in a niche-dependent manner. In summary, MBNL3 down-regulation-related reversion to an embryonic alternative splicing program, typified by CD44v3 overexpression, represents a previously unidentified mechanism governing malignant progenitor reprogramming in malignant microenvironments and provides a pivotal opportunity for selective BC LSC detection and therapeutic elimination.


Subject(s)
Alternative Splicing/genetics , Cell Self Renewal/genetics , Human Embryonic Stem Cells/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Adult , Animals , Apoptosis/genetics , Blast Crisis/genetics , Blast Crisis/pathology , Bone Marrow/pathology , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Proliferation , Cell Survival , Cellular Reprogramming/genetics , Female , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Leukemic , Gene Knockdown Techniques , Hematopoiesis , Humans , Hyaluronan Receptors/metabolism , Ligands , Male , Mice , Middle Aged , Neoplasm Transplantation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pluripotent Stem Cells/cytology , Proto-Oncogene Mas
SELECTION OF CITATIONS
SEARCH DETAIL
...