Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Ann Surg ; 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38887930

ABSTRACT

OBJECTIVE: To assess the utility of tumor-intrinsic and cancer-associated fibroblast (CAF) subtypes of pancreatic ductal adenocarcinoma (PDAC) in predicting response to neoadjuvant therapy (NAT) and overall survival. BACKGROUND: PDAC remains a deadly disease with limited treatment options, and both the tumor as well as the microenvironment play an important role in pathogenesis. Gene expression-based tumor-intrinsic subtypes (classical and basal-like) have been shown to predict outcomes, but tumor microenvironment subtypes are still evolving. METHODS: RNA-sequencing was performed on 114 deidentified resected PDAC tumors. Clinical data were collected by retrospective chart review. Single sample classifiers (SSCs) were used to determine classical and basal-like subtypes as well as tumor-permissive permCAF and tumor-restraining restCAF subtypes. Survival was analyzed using log-rank test. RESULTS: Patients who received NAT had an increase in overall survival (OS), with median survival of 27.9 months compared to 20.1 months for those who did not receive NAT, but the difference did not reach statistical significance (HR 0.64, P=0.076). Either tumor-intrinsic or CAF subtypes alone were associated with OS regardless of NAT or no NAT, and patients with classical or restCAF subtype had the best outcomes. When evaluated together, patients with classical-restCAF subtype had the best OS and basal-permCAF the worst OS (P<0.0001). NAT patients with classical-restCAF subtype demonstrated the longest OS compared to the other groups (P=0.00041). CONCLUSIONS: CAF subtypes have an additive effect over tumor-intrinsic subtypes in predicting survival with or without neoadjuvant FOLFIRINOX in PDAC. Molecular subtyping of both tumor and CAF compartments of PDAC may be important steps in selecting first-line systemic therapy.

2.
bioRxiv ; 2024 May 17.
Article in English | MEDLINE | ID: mdl-38798565

ABSTRACT

Cancer-associated fibroblast (CAF) subpopulations in pancreatic ductal adenocarcinoma (PDAC) have been identified using single-cell RNA sequencing (scRNAseq) with divergent characteristics, but their clinical relevance remains unclear. We translate scRNAseq-derived CAF cell-subpopulation-specific marker genes to bulk RNAseq data, and develop a single- sample classifier, DeCAF, for the classification of clinically rest raining and perm issive CAF subtypes. We validate DeCAF in 19 independent bulk transcriptomic datasets across four tumor types (PDAC, mesothelioma, bladder and renal cell carcinoma). DeCAF subtypes have distinct histology features, immune landscapes, and are prognostic and predict response to therapy across cancer types. We demonstrate that DeCAF is clinically replicable and robust for the classification of CAF subtypes in patients for multiple tumor types, providing a better framework for the future development and translation of therapies against permissive CAF subtypes and preservation of restraining CAF subtypes. Significance: We introduce a replicable and robust classifier, DeCAF, that delineates the significance of the role of permissive and restraining CAF subtypes in cancer patients. DeCAF is clinically tractable, prognostic and predictive of treatment response in multiple cancer types and lays the translational groundwork for the preclinical and clinical development of CAF subtype specific therapies.

3.
Biometrics ; 80(1)2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38497825

ABSTRACT

Modern biomedical datasets are increasingly high-dimensional and exhibit complex correlation structures. Generalized linear mixed models (GLMMs) have long been employed to account for such dependencies. However, proper specification of the fixed and random effects in GLMMs is increasingly difficult in high dimensions, and computational complexity grows with increasing dimension of the random effects. We present a novel reformulation of the GLMM using a factor model decomposition of the random effects, enabling scalable computation of GLMMs in high dimensions by reducing the latent space from a large number of random effects to a smaller set of latent factors. We also extend our prior work to estimate model parameters using a modified Monte Carlo Expectation Conditional Minimization algorithm, allowing us to perform variable selection on both the fixed and random effects simultaneously. We show through simulation that through this factor model decomposition, our method can fit high-dimensional penalized GLMMs faster than comparable methods and more easily scale to larger dimensions not previously seen in existing approaches.


Subject(s)
Algorithms , Computer Simulation , Linear Models , Monte Carlo Method
4.
J Surg Oncol ; 129(5): 860-868, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38233984

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) has a fibrotic stroma that has both tumor-promoting and tumor-restraining properties. Different types of cancer-associated fibroblasts (CAFs) have been described. Here, we investigated whether CAFs within the same subtype exhibit heterogeneous functions. METHODS: We evaluated the gene and protein expression differences in two myofibroblastic CAF (myCAF) lines using single-cell and bulk RNA-sequencing. We utilized proliferation and migration assays to determine the effect of different CAF lines on a tumor cell line. RESULTS: We found that myCAF lines express an activated stroma subtype gene signature, which is associated with a shorter survival in patients. Although both myCAF lines expressed α-smooth muscle actin (α-SMA), platelet-derived growth factor-α (PDGFR-α), fibroblast-activated protein (FAP), and vimentin, we observed heterogeneity between the two lines. Similarly, despite being consistent with myCAF gene expression overall, heterogeneity within specific genes was observed. We found that these differences extended to the functional level where the two myCAF lines had different effects on the same tumor cell line. The myCAF216 line, which had slightly increased inflammatory CAF-like gene expression and higher protein expression of α-SMA, PDGFR-α, and FAP was found to restrain migration of tumor cells. CONCLUSIONS: We found that two myCAF lines with globally similar expression characteristics had different effects on the same tumor cell line, one promoting and the other restraining migration. Our study highlights that there may be unappreciated heterogeneity within CAF subtypes. Further investigation and attention to specific genes or proteins that may drive this heterogeneity will be important.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Fibroblasts/metabolism , Cell Line, Tumor , Tumor Microenvironment
5.
Bioinformatics ; 39(8)2023 08 01.
Article in English | MEDLINE | ID: mdl-37498558

ABSTRACT

MOTIVATION: Single-cell RNA-sequencing (scRNA-seq) has enabled the molecular profiling of thousands to millions of cells simultaneously in biologically heterogenous samples. Currently, the common practice in scRNA-seq is to determine cell type labels through unsupervised clustering and the examination of cluster-specific genes. However, even small differences in analysis and parameter choosing can greatly alter clustering results and thus impose great influence on which cell types are identified. Existing methods largely focus on determining the optimal number of robust clusters, which can be problematic for identifying cells of extremely low abundance due to their subtle contributions toward overall patterns of gene expression. RESULTS: Here, we present a carefully designed framework, SCISSORS, which accurately profiles subclusters within broad cluster(s) for the identification of rare cell types in scRNA-seq data. SCISSORS employs silhouette scoring for the estimation of heterogeneity of clusters and reveals rare cells in heterogenous clusters by a multi-step semi-supervised reclustering process. Additionally, SCISSORS provides a method for the identification of marker genes of high specificity to the cell type. SCISSORS is wrapped around the popular Seurat R package and can be easily integrated into existing Seurat pipelines. AVAILABILITY AND IMPLEMENTATION: SCISSORS, including source code and vignettes, are freely available at https://github.com/jr-leary7/SCISSORS.


Subject(s)
Algorithms , Gene Expression Profiling , Gene Expression Profiling/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Cluster Analysis , RNA
6.
Commun Biol ; 6(1): 163, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36765128

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive disease for which potent therapies have limited efficacy. Several studies have described the transcriptomic landscape of PDAC tumors to provide insight into potentially actionable gene expression signatures to improve patient outcomes. Despite centralization efforts from multiple organizations and increased transparency requirements from funding agencies and publishers, analysis of public PDAC data remains difficult. Bioinformatic pitfalls litter public transcriptomic data, such as subtle inclusion of low-purity and non-adenocarcinoma cases. These pitfalls can introduce non-specificity to gene signatures without appropriate data curation, which can negatively impact findings. To reduce barriers to analysis, we have created pdacR ( http://pdacR.bmi.stonybrook.edu , github.com/rmoffitt/pdacR), an open-source software package and web-tool with annotated datasets from landmark studies and an interface for user-friendly analysis in clustering, differential expression, survival, and dimensionality reduction. Using this tool, we present a multi-dataset analysis of PDAC transcriptomics that confirms the basal-like/classical model over alternatives.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Prognosis , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Gene Expression Profiling , Pancreatic Neoplasms
7.
Int J Mol Sci ; 23(10)2022 May 13.
Article in English | MEDLINE | ID: mdl-35628269

ABSTRACT

Elevated levels of Mucin-16 (MUC16) in conjunction with a high expression of truncated O-glycans is implicated in playing crucial roles in the malignancy of pancreatic ductal adenocarcinoma (PDAC). However, the mechanisms by which such aberrant glycoforms present on MUC16 itself promote an increased disease burden in PDAC are yet to be elucidated. This study demonstrates that the CRISPR/Cas9-mediated genetic deletion of MUC16 in PDAC cells decreases tumor cell migration. We found that MUC16 enhances tumor malignancy by activating the integrin-linked kinase and focal adhesion kinase (ILK/FAK)-signaling axis. These findings are especially noteworthy in truncated O-glycan (Tn and STn antigen)-expressing PDAC cells. Activation of these oncogenic-signaling pathways resulted in part from interactions between MUC16 and integrin complexes (α4ß1), which showed a stronger association with aberrant glycoforms of MUC16. Using a monoclonal antibody to functionally hinder MUC16 significantly reduced the migratory cascades in our model. Together, these findings suggest that truncated O-glycan containing MUC16 exacerbates malignancy in PDAC by activating FAK signaling through specific interactions with α4 and ß1 integrin complexes on cancer cell membranes. Targeting these aberrant glycoforms of MUC16 can aid in the development of a novel platform to study and treat metastatic pancreatic cancer.


Subject(s)
CA-125 Antigen , Carcinoma, Pancreatic Ductal , Focal Adhesion Kinase 1 , Integrin alpha4beta1 , Membrane Proteins , Pancreatic Neoplasms , CA-125 Antigen/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Focal Adhesion Kinase 1/metabolism , Humans , Integrin alpha4beta1/metabolism , Membrane Proteins/metabolism , Pancreatic Hormones/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Polysaccharides/metabolism
8.
Stem Cell Reports ; 16(10): 2442-2458, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34534448

ABSTRACT

Skeletal muscle satellite cells (SCs) are stem cells responsible for muscle development and regeneration. Although CRISPR/Cas9 has been widely used, its application in endogenous SCs remains elusive. Here, we generate mice expressing Cas9 in SCs and achieve robust editing in juvenile SCs at the postnatal stage through AAV9-mediated short guide RNA (sgRNA) delivery. Additionally, we reveal that quiescent SCs are resistant to CRISPR/Cas9-mediated editing. As a proof of concept, we demonstrate efficient editing of master transcription factor (TF) Myod1 locus using the CRISPR/Cas9/AAV9-sgRNA system in juvenile SCs. Application on two key TFs, MYC and BCL6, unveils distinct functions in SC activation and muscle regeneration. Particularly, we reveal that MYC orchestrates SC activation through regulating 3D genome architecture. Its depletion results in strengthening of the topologically associating domain boundaries thus may affect gene expression. Altogether, our study establishes a platform for editing endogenous SCs that can be harnessed to elucidate the functionality of key regulators governing SC activities.


Subject(s)
Chromatin/metabolism , Genes, myc , Genome , MyoD Protein/metabolism , Proto-Oncogene Proteins c-bcl-6/metabolism , RNA, Guide, Kinetoplastida/metabolism , Satellite Cells, Skeletal Muscle/physiology , Animals , CRISPR-Cas Systems , Gene Editing/methods , Gene Expression Regulation , Mice , MyoD Protein/genetics , Nucleic Acid Conformation , Proto-Oncogene Proteins c-bcl-6/genetics , RNA, Guide, Kinetoplastida/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Elife ; 102021 05 19.
Article in English | MEDLINE | ID: mdl-34009124

ABSTRACT

To study disease development, an inventory of an organ's cell types and understanding of physiologic function is paramount. Here, we performed single-cell RNA-sequencing to examine heterogeneity of murine pancreatic duct cells, pancreatobiliary cells, and intrapancreatic bile duct cells. We describe an epithelial-mesenchymal transitory axis in our three pancreatic duct subpopulations and identify osteopontin as a regulator of this fate decision as well as human duct cell dedifferentiation. Our results further identify functional heterogeneity within pancreatic duct subpopulations by elucidating a role for geminin in accumulation of DNA damage in the setting of chronic pancreatitis. Our findings implicate diverse functional roles for subpopulations of pancreatic duct cells in maintenance of duct cell identity and disease progression and establish a comprehensive road map of murine pancreatic duct cell, pancreatobiliary cell, and intrapancreatic bile duct cell homeostasis.


Subject(s)
Gene Expression Profiling , Genetic Heterogeneity , Pancreatic Ducts/cytology , Single-Cell Analysis , Transcriptome , Animals , Cell Line , Cell Separation , DNA Damage , Databases, Genetic , Disease Models, Animal , Epithelial-Mesenchymal Transition , Female , Geminin/genetics , Geminin/metabolism , Gene Expression Regulation, Developmental , Humans , Mice, Inbred C57BL , Mice, Transgenic , Morphogenesis , Osteopontin/genetics , Osteopontin/metabolism , Pancreatic Ducts/metabolism , Pancreatitis, Chronic/genetics , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Phenotype , RNA-Seq
10.
Mol Ther ; 29(4): 1557-1571, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33359791

ABSTRACT

Aberrant expression of CA125/MUC16 is associated with pancreatic ductal adenocarcinoma (PDAC) progression and metastasis. However, knowledge of the contribution of MUC16 to pancreatic tumorigenesis is limited. Here, we show that MUC16 expression is associated with disease progression, basal-like and squamous tumor subtypes, increased tumor metastasis, and short-term survival of PDAC patients. MUC16 enhanced tumor malignancy through the activation of AKT and GSK3ß oncogenic signaling pathways. Activation of these oncogenic signaling pathways resulted in part from increased interactions between MUC16 and epidermal growth factor (EGF)-type receptors, which were enhanced for aberrant glycoforms of MUC16. Treatment of PDAC cells with monoclonal antibody (mAb) AR9.6 significantly reduced MUC16-induced oncogenic signaling. mAb AR9.6 binds to a unique conformational epitope on MUC16, which is influenced by O-glycosylation. Additionally, treatment of PDAC tumor-bearing mice with either mAb AR9.6 alone or in combination with gemcitabine significantly reduced tumor growth and metastasis. We conclude that the aberrant expression of MUC16 enhances PDAC progression to an aggressive phenotype by modulating oncogenic signaling through ErbB receptors. Anti-MUC16 mAb AR9.6 blocks oncogenic activities and tumor growth and could be a novel immunotherapeutic agent against MUC16-mediated PDAC tumor malignancy.


Subject(s)
Adenocarcinoma/drug therapy , CA-125 Antigen/genetics , Carcinogenesis/genetics , Carcinoma, Pancreatic Ductal/drug therapy , ErbB Receptors/genetics , Membrane Proteins/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Antibodies, Monoclonal/pharmacology , CA-125 Antigen/immunology , Carcinogenesis/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/immunology , Mice , Neoplasm Metastasis , Protein Isoforms/genetics , Protein Isoforms/immunology , Signal Transduction
11.
JCI Insight ; 5(8)2020 04 23.
Article in English | MEDLINE | ID: mdl-32213714

ABSTRACT

Over 55,000 people in the United States are diagnosed with pancreatic ductal adenocarcinoma (PDAC) yearly, and fewer than 20% of these patients survive a year beyond diagnosis. Chemotherapies are considered or used in nearly every PDAC case, but there is limited understanding of the complex signaling responses underlying resistance to these common treatments. Here, we take an unbiased approach to study protein kinase network changes following chemotherapies in patient-derived xenograft (PDX) models of PDAC to facilitate design of rational drug combinations. Proteomics profiling following chemotherapy regimens reveals that activation of JNK-JUN signaling occurs after 5-fluorouracil plus leucovorin (5-FU + LEU) and FOLFOX (5-FU + LEU plus oxaliplatin [OX]), but not after OX alone or gemcitabine. Cell and tumor growth assays with the irreversible inhibitor JNK-IN-8 and genetic manipulations demonstrate that JNK and JUN each contribute to chemoresistance and cancer cell survival after FOLFOX. Active JNK1 and JUN are specifically implicated in these effects, and synergy with JNK-IN-8 is linked to FOLFOX-mediated JUN activation, cell cycle dysregulation, and DNA damage response. This study highlights the potential for JNK-IN-8 as a biological tool and potential combination therapy with FOLFOX in PDAC and reinforces the need to tailor treatment to functional characteristics of individual tumors.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Pancreatic Ductal , Drug Resistance, Neoplasm/drug effects , Pancreatic Neoplasms , Animals , Antineoplastic Combined Chemotherapy Protocols , Drug Evaluation, Preclinical , Fluorouracil/pharmacology , Humans , Leucovorin , MAP Kinase Kinase 4/antagonists & inhibitors , Mice , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , Organoplatinum Compounds , Xenograft Model Antitumor Assays
12.
Clin Cancer Res ; 26(1): 82-92, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31754050

ABSTRACT

PURPOSE: Molecular subtyping for pancreatic cancer has made substantial progress in recent years, facilitating the optimization of existing therapeutic approaches to improve clinical outcomes in pancreatic cancer. With advances in treatment combinations and choices, it is becoming increasingly important to determine ways to place patients on the best therapies upfront. Although various molecular subtyping systems for pancreatic cancer have been proposed, consensus regarding proposed subtypes, as well as their relative clinical utility, remains largely unknown and presents a natural barrier to wider clinical adoption. EXPERIMENTAL DESIGN: We assess three major subtype classification schemas in the context of results from two clinical trials and by meta-analysis of publicly available expression data to assess statistical criteria of subtype robustness and overall clinical relevance. We then developed a single-sample classifier (SSC) using penalized logistic regression based on the most robust and replicable schema. RESULTS: We demonstrate that a tumor-intrinsic two-subtype schema is most robust, replicable, and clinically relevant. We developed Purity Independent Subtyping of Tumors (PurIST), a SSC with robust and highly replicable performance on a wide range of platforms and sample types. We show that PurIST subtypes have meaningful associations with patient prognosis and have significant implications for treatment response to FOLIFIRNOX. CONCLUSIONS: The flexibility and utility of PurIST on low-input samples such as tumor biopsies allows it to be used at the time of diagnosis to facilitate the choice of effective therapies for patients with pancreatic ductal adenocarcinoma and should be considered in the context of future clinical trials.


Subject(s)
Biomarkers, Tumor/genetics , Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Molecular Typing/methods , Pancreatic Neoplasms/classification , Pancreatic Neoplasms/pathology , Clinical Trials as Topic/statistics & numerical data , Databases, Genetic/statistics & numerical data , Humans , Pancreatic Neoplasms/genetics , Survival Rate , Treatment Outcome
13.
Nat Commun ; 10(1): 4729, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31628300

ABSTRACT

Tumors are mixtures of different compartments. While global gene expression analysis profiles the average expression of all compartments in a sample, identifying the specific contribution of each compartment remains a challenge. With the increasing recognition of the importance of non-neoplastic components, the ability to breakdown the gene expression contribution of each is critical. Here, we develop DECODER, an integrated framework which performs de novo deconvolution and single-sample compartment weight estimation. We use DECODER to deconvolve 33 TCGA tumor RNA-seq data sets and show that it may be applied to other data types including ATAC-seq. We demonstrate that it can be utilized to reproducibly estimate cellular compartment weights in pancreatic cancer that are clinically meaningful. Application of DECODER across cancer types advances the capability of identifying cellular compartments in an unknown sample and may have implications for identifying the tumor of origin for cancers of unknown primary.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Algorithms , Humans , Models, Genetic , Neoplasms/classification , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Reproducibility of Results , Sequence Analysis, RNA , Software , Tumor Burden/genetics
14.
Methods Mol Biol ; 1909: 177-180, 2019.
Article in English | MEDLINE | ID: mdl-30580431

ABSTRACT

The discovery of circulating cell-free fetal DNA has profoundly transformed the landscape of noninvasive prenatal testing (NIPT) and rapidly found its way into global clinical applications. The fractional concentration of cell-free fetal DNA in plasma DNA of a pregnant woman is an important parameter for understanding and interpreting analytical results of NIPT. Thus, the accurate quantification of fetal DNA fraction is indispensable when NIPT is involved. In this protocol, we describe the bioinformatics workflow to calculate fetal DNA fraction using two programs developed by our group, which provide accurate estimation.


Subject(s)
Cell-Free Nucleic Acids/genetics , Fetus/metabolism , Genomics/methods , Prenatal Diagnosis/methods , Software , Cell-Free Nucleic Acids/analysis , Cell-Free Nucleic Acids/blood , Female , Genetic Testing/methods , Humans , Pregnancy , Workflow
16.
Methods Mol Biol ; 1668: 15-25, 2017.
Article in English | MEDLINE | ID: mdl-28842899

ABSTRACT

Transcriptional control of gene expression in skeletal muscle cell is involved in different processes ranging from muscle formation to regeneration. The identification of an increasing number of transcription factors, co-factors, and histone modifications has been greatly advanced by methods that allow studies of genome-wide chromatin-protein interactions. Chromatin immunoprecipitation with massively parallel DNA sequencing, or ChIP-seq, is a powerful tool for identifying binding sites of TFs/co-factors and histone modifications. The major steps of this technique involve immunoprecipitation of fragmented chromatin, followed by high-throughput sequencing to identify the protein bound regions genome-wide. Here, in this protocol, we will illustrate how the entire ChIP-seq is performed using global H3K27ac profiling in myoblast cells as an example.


Subject(s)
Chromatin Immunoprecipitation/methods , Chromatin/genetics , Muscle Fibers, Skeletal/metabolism , Transcription Factors/metabolism , Whole Genome Sequencing/methods , Animals , Binding Sites/genetics , Cell Line , Chromosome Mapping , Gene Library , Genome-Wide Association Study , Histones/genetics , Histones/metabolism , Mice , Transcription Factors/genetics
17.
Nucleic Acids Res ; 45(15): 8785-8805, 2017 Sep 06.
Article in English | MEDLINE | ID: mdl-28575289

ABSTRACT

Super-enhancers (SEs) are cis-regulatory elements enriching lineage specific key transcription factors (TFs) to form hotspots. A paucity of identification and functional dissection promoted us to investigate SEs during myoblast differentiation. ChIP-seq analysis of histone marks leads to the uncovering of SEs which remodel progressively during the course of differentiation. Further analyses of TF ChIP-seq enable the definition of SE hotspots co-bound by the master TF, MyoD and other TFs, among which we perform in-depth dissection for MyoD/FoxO3 interaction in driving the hotspots formation and SE activation. Furthermore, using Myogenin as a model locus, we elucidate the hierarchical and complex interactions among hotspots during the differentiation, demonstrating SE function is propelled by the physical and functional cooperation among hotspots. Finally, we show MyoD and FoxO3 are key in orchestrating the Myogenin hotspots interaction and activation. Altogether our results identify muscle-specific SEs and provide mechanistic insights into the functionality of SE.


Subject(s)
Cell Differentiation/genetics , Enhancer Elements, Genetic/physiology , Forkhead Box Protein O3/physiology , Muscle Development/genetics , MyoD Protein/physiology , Animals , Cells, Cultured , Forkhead Box Protein O3/metabolism , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Mice , MyoD Protein/metabolism , Myoblasts/physiology , Myogenin/genetics , Myogenin/metabolism , Protein Binding
18.
Methods Mol Biol ; 1556: 355-362, 2017.
Article in English | MEDLINE | ID: mdl-28247361

ABSTRACT

Long intergenic noncoding RNAs (lincRNAs) have emerged as critical participators in gene regulation in myriads of cell types. The development of the whole transcriptome sequencing technology, or RNA-seq , has enabled novel lincRNA detection, but the bioinformatics analysis toward distinguishing reliable ones remains a challenge. Here, we describe the bioinformatics workflow developed for identifying novel lincRNAs step by step, including read alignment, transcriptome assembly and transcript filtering.


Subject(s)
Computational Biology/methods , Gene Expression Profiling/methods , Muscle Fibers, Skeletal/metabolism , Databases, Nucleic Acid , Genomics/methods , Humans , RNA, Long Noncoding , Software , Transcriptome , Web Browser , Workflow
19.
Int J Mol Sci ; 18(2)2017 Feb 20.
Article in English | MEDLINE | ID: mdl-28230760

ABSTRACT

The discovery of cell-free fetal DNA molecules in plasma of pregnant women has created a paradigm shift in noninvasive prenatal testing (NIPT). Circulating cell-free DNA in maternal plasma has been increasingly recognized as an important proxy to detect fetal abnormalities in a noninvasive manner. A variety of approaches for NIPT using next-generation sequencing have been developed, which have been rapidly transforming clinical practices nowadays. In such approaches, the fetal DNA fraction is a pivotal parameter governing the overall performance and guaranteeing the proper clinical interpretation of testing results. In this review, we describe the current bioinformatics approaches developed for estimating the fetal DNA fraction and discuss their pros and cons.


Subject(s)
Computational Biology , DNA/genetics , Fetus , Genetic Testing , Prenatal Diagnosis , Chromosomes, Human, Y/genetics , Computational Biology/methods , DNA/blood , DNA Methylation , Female , Genetic Testing/methods , Genotype , High-Throughput Nucleotide Sequencing , Humans , Molecular Typing , Polymorphism, Single Nucleotide , Pregnancy , Prenatal Diagnosis/methods , Sequence Analysis, DNA
20.
NPJ Genom Med ; 1: 16013, 2016.
Article in English | MEDLINE | ID: mdl-29263813

ABSTRACT

Noninvasive prenatal testing using massively parallel sequencing of maternal plasma DNA has been rapidly adopted in clinical use worldwide. Fetal DNA fraction in a maternal plasma sample is an important parameter for accurate interpretations of these tests. However, there is a lack of methods involving low-sequencing depth and yet would allow a robust and accurate determination of fetal DNA fraction in maternal plasma for all pregnancies. In this study, we have developed a new method to accurately quantify the fetal DNA fraction by analysing the maternal genotypes and sequencing data of maternal plasma DNA. Fetal DNA fraction was calculated based on the proportion of non-maternal alleles at single-nucleotide polymorphisms where the mother is homozygous. This new approach achieves a median deviation of 0.6% between predicted fetal DNA fraction and the actual fetal DNA fraction using as low as 0.03-fold sequencing coverage of the human genome. We believe that this method will further enhance the clinical interpretations of noninvasive prenatal testing using genome-wide random sequencing.

SELECTION OF CITATIONS
SEARCH DETAIL
...