Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cell Genom ; 4(7): 100591, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38925123

ABSTRACT

Understanding the complex interplay of genetic and environmental factors in disease etiology and the role of gene-environment interactions (GEIs) across human development stages is important. We review the state of GEI research, including challenges in measuring environmental factors and advantages of GEI analysis in understanding disease mechanisms. We discuss the evolution of GEI studies from candidate gene-environment studies to genome-wide interaction studies (GWISs) and the role of multi-omics in mediating GEI effects. We review advancements in GEI analysis methods and the importance of large-scale datasets. We also address the translation of GEI findings into precision environmental health (PEH), showcasing real-world applications in healthcare and disease prevention. Additionally, we highlight societal considerations in GEI research, including environmental justice, the return of results to participants, and data privacy. Overall, we underscore the significance of GEI for disease prediction and prevention and advocate for integrating the exposome into PEH omics studies.


Subject(s)
Environmental Health , Gene-Environment Interaction , Precision Medicine , Humans , Precision Medicine/methods , Genome-Wide Association Study , Environmental Exposure/adverse effects
2.
Brief Bioinform ; 25(1)2023 11 22.
Article in English | MEDLINE | ID: mdl-38113074

ABSTRACT

Optimizing and benchmarking data reduction methods for dynamic or spatial visualization and interpretation (DSVI) face challenges due to many factors, including data complexity, lack of ground truth, time-dependent metrics, dimensionality bias and different visual mappings of the same data. Current studies often focus on independent static visualization or interpretability metrics that require ground truth. To overcome this limitation, we propose the MIBCOVIS framework, a comprehensive and interpretable benchmarking and computational approach. MIBCOVIS enhances the visualization and interpretability of high-dimensional data without relying on ground truth by integrating five robust metrics, including a novel time-ordered Markov-based structural metric, into a semi-supervised hierarchical Bayesian model. The framework assesses method accuracy and considers interaction effects among metric features. We apply MIBCOVIS using linear and nonlinear dimensionality reduction methods to evaluate optimal DSVI for four distinct dynamic and spatial biological processes captured by three single-cell data modalities: CyTOF, scRNA-seq and CODEX. These data vary in complexity based on feature dimensionality, unknown cell types and dynamic or spatial differences. Unlike traditional single-summary score approaches, MIBCOVIS compares accuracy distributions across methods. Our findings underscore the joint evaluation of visualization and interpretability, rather than relying on separate metrics. We reveal that prioritizing average performance can obscure method feature performance. Additionally, we explore the impact of data complexity on visualization and interpretability. Specifically, we provide optimal parameters and features and recommend methods, like the optimized variational contractive autoencoder, for targeted DSVI for various data complexities. MIBCOVIS shows promise for evaluating dynamic single-cell atlases and spatiotemporal data reduction models.


Subject(s)
Benchmarking , Single-Cell Analysis , Bayes Theorem , Single-Cell Analysis/methods
3.
Brief Bioinform ; 23(2)2022 03 10.
Article in English | MEDLINE | ID: mdl-35192692

ABSTRACT

A major topic of debate in developmental biology centers on whether development is continuous, discontinuous, or a mixture of both. Pseudo-time trajectory models, optimal for visualizing cellular progression, model cell transitions as continuous state manifolds and do not explicitly model real-time, complex, heterogeneous systems and are challenging for benchmarking with temporal models. We present a data-driven framework that addresses these limitations with temporal single-cell data collected at discrete time points as inputs and a mixture of dependent minimum spanning trees (MSTs) as outputs, denoted as dynamic spanning forest mixtures (DSFMix). DSFMix uses decision-tree models to select genes that account for variations in multimodality, skewness and time. The genes are subsequently used to build the forest using tree agglomerative hierarchical clustering and dynamic branch cutting. We first motivate the use of forest-based algorithms compared to single-tree approaches for visualizing and characterizing developmental processes. We next benchmark DSFMix to pseudo-time and temporal approaches in terms of feature selection, time correlation, and network similarity. Finally, we demonstrate how DSFMix can be used to visualize, compare and characterize complex relationships during biological processes such as epithelial-mesenchymal transition, spermatogenesis, stem cell pluripotency, early transcriptional response from hormones and immune response to coronavirus disease. Our results indicate that the expression of genes during normal development exhibits a high proportion of non-uniformly distributed profiles that are mostly right-skewed and multimodal; the latter being a characteristic of major steady states during development. Our study also identifies and validates gene signatures driving complex dynamic processes during somatic or germline differentiation.


Subject(s)
Benchmarking , Models, Theoretical , Single-Cell Analysis/methods , Algorithms , Animals , Cellular Microenvironment , Data Analysis , Decision Trees , Gene Expression Profiling/methods , Humans , Spermatogenesis
4.
Front Mol Biosci ; 9: 777390, 2022.
Article in English | MEDLINE | ID: mdl-35155574

ABSTRACT

During an adaptive immune response from pathogen invasion, multiple cytokines are produced by various immune cells interacting jointly at the cellular level to mediate several processes. For example, studies have shown that regulation of interleukin-4 (IL-4) correlates with interleukin-2 (IL-2) induced lymphocyte proliferation. This motivates the need to better understand and model the mechanisms driving the dynamic interplay of proliferation of lymphocytes with the complex interaction effects of cytokines during an immune response. To address this challenge, we adopt a hybrid computational approach comprising of continuous, discrete and stochastic non-linear model formulations to predict a system-level immune response as a function of multiple dependent signals and interacting agents including cytokines and targeted immune cells. We propose a hybrid ordinary differential equation-based (ODE) multicellular model system with a stochastic component of antigen microscopic states denoted as Multiscale Multicellular Quantitative Evaluator (MMQE) implemented using MATLAB. MMQE combines well-defined immune response network-based rules and ODE models to capture the complex dynamic interactions between the proliferation levels of different types of communicating lymphocyte agents mediated by joint regulation of IL-2 and IL-4 to predict the emergent global behavior of the system during an immune response. We model the activation of the immune system in terms of different activation protocols of helper T cells by the interplay of independent biological agents of classic antigen-presenting cells (APCs) and their joint activation which is confounded by the exposure time to external pathogens. MMQE quantifies the dynamics of lymphocyte proliferation during pathogen invasion as bivariate distributions of IL-2 and IL-4 concentration levels. Specifically, by varying activation agents such as dendritic cells (DC), B cells and their joint mechanism of activation, we quantify how lymphocyte activation and differentiation protocols boost the immune response against pathogen invasion mediated by a joint downregulation of IL-4 and upregulation of IL-2. We further compare our in-silico results to in-vivo and in-vitro experimental studies for validation. In general, MMQE combines intracellular and extracellular effects from multiple interacting systems into simpler dynamic behaviors for better interpretability. It can be used to aid engineering of anti-infection drugs or optimizing drug combination therapies against several diseases.

5.
PLoS One ; 17(1): e0262639, 2022.
Article in English | MEDLINE | ID: mdl-35061813

ABSTRACT

One important metric of a radiologist's visibility and influence is their ability to participate in discussion within their community. The goal of our study was to compare the participation level of men and women in scientific discussions at the annual meeting of the Radiological Society of North America (RSNA). Eleven volunteers collected participation data by gender in 59 sessions (286 presentations) at the 2018 RSNA meeting. Data was analyzed using a combination of Chi-squared, paired Wilcoxon signed-rank and T-test. Of all RSNA professional attendees at the RSNA, 68% were men and 32% were women. Of the 2869 presentations listed in the program, 65% were presented by men and 35% were presented by women. Of the 286 presentations in our sample, 177 (61.8%) were presented by men and 109 (38.1%) were presented by women. Of these 286 presentations, 81 (63%) were moderated by men and 47 (37%) were moderated by women. From the audience, 190 male attendees participated in 134 question-and-answer (Q&A) sessions following presentations and 58 female attendees participated in 52 Q&A sessions (P<0.001). Female attendees who did participate in Q&A sessions talked for a significantly shorter period of time (mean 7.14 ± 17.7 seconds, median 0) compared to male attendees (28.7 ± 29.6 seconds, median 16; P<0.001). Overall, our findings demonstrate that women participated less than men in the Q&A sessions at RSNA 2018, and talked for a shorter period of time. The fact that women were outnumbered among their male peers may explain the difference in behavior by gender.


Subject(s)
Congresses as Topic/statistics & numerical data , Radiologists/statistics & numerical data , Sexism/statistics & numerical data , Career Mobility , Female , Humans , Male , Radiology/statistics & numerical data , Sex Factors
6.
Sci Rep ; 12(1): 1393, 2022 01 26.
Article in English | MEDLINE | ID: mdl-35082309

ABSTRACT

The interplay between genes harboring single nucleotide polymorphisms (SNPs) is vital to better understand underlying contributions to the etiology of breast cancer. Much attention has been paid to epistasis between nuclear genes or mutations in the mitochondrial genome. However, there is limited understanding about the epistatic effects of genetic variants in the nuclear and mitochondrial genomes jointly on breast cancer. We tested the interaction of germline SNPs in the mitochondrial (mtSNPs) and nuclear (nuSNPs) genomes of female breast cancer patients in The Cancer Genome Atlas (TCGA) for association with morphological features extracted from hematoxylin and eosin (H&E)-stained pathology images. We identified 115 significant (q-value < 0.05) mito-nuclear interactions that increased nuclei size by as much as 12%. One interaction between nuSNP rs17320521 in an intron of the WSC Domain Containing 2 (WSCD2) gene and mtSNP rs869096886, a synonymous variant mapped to the mitochondrially-encoded NADH dehydrogenase 4 (MT-ND4) gene, was confirmed in an independent breast cancer data set from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC). None of the 10 mito-nuclear interactions identified from non-diseased female breast tissues from the Genotype-Expression (GTEx) project resulted in an increase in nuclei size. Comparisons of gene expression data from the TCGA breast cancer patients with the genotype homozygous for the minor alleles of the SNPs in WSCD2 and MT-ND4 versus the other genotypes revealed core transcriptional regulator interactions and an association with insulin. Finally, a Cox proportional hazards ratio = 1.7 (C.I. 0.98-2.9, p-value = 0.042) and Kaplan-Meier plot suggest that the TCGA female breast cancer patients with low gene expression of WSCD2 coupled with large nuclei have an increased risk of mortality. The intergenomic dependency between the two variants may constitute an inherent susceptibility of a more severe form of breast cancer and points to genetic targets for further investigation of additional determinants of the disease.


Subject(s)
Biological Variation, Population/genetics , Breast Neoplasms/genetics , Cell Nucleus/genetics , Epistasis, Genetic , Genome, Mitochondrial , Mitochondria/genetics , Polymorphism, Single Nucleotide , Alleles , Cell Communication/genetics , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cell Size , Female , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , Genetic Predisposition to Disease , Homozygote , Humans , Introns , Mitochondria/metabolism
7.
Pharmacogenomics ; 22(9): 543-551, 2021 06.
Article in English | MEDLINE | ID: mdl-34044623

ABSTRACT

Combination drug therapies have become an integral part of precision oncology, and while evidence of clinical effectiveness continues to grow, the underlying mechanisms supporting synergy are poorly understood. Immortalized human lymphoblastoid cell lines (LCLs) have been proven as a particularly useful, scalable and low-cost model in pharmacogenetics research, and are suitable for elucidating the molecular mechanisms of synergistic combination therapies. In this review, we cover the advantages of LCLs in synergy pharmacogenomics and consider recent studies providing initial evidence of the utility of LCLs in synergy research. We also discuss several opportunities for LCL-based systems to address gaps in the research through the expansion of testing regimens, assessment of new drug classes and higher-order combinations, and utilization of integrated omics technologies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor/drug effects , Lymphocytes/drug effects , Pharmacogenomic Testing/methods , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Humans
8.
Nature ; 588(7839): 670-675, 2020 12.
Article in English | MEDLINE | ID: mdl-33238290

ABSTRACT

The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange. Three-dimensional in vitro human distal lung culture systems would strongly facilitate the investigation of pathologies such as interstitial lung disease, cancer and coronavirus disease 2019 (COVID-19) pneumonia caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we describe the development of a long-term feeder-free, chemically defined culture system for distal lung progenitors as organoids derived from single adult human alveolar epithelial type II (AT2) or KRT5+ basal cells. AT2 organoids were able to differentiate into AT1 cells, and basal cell organoids developed lumens lined with differentiated club and ciliated cells. Single-cell analysis of KRT5+ cells in basal organoids revealed a distinct population of ITGA6+ITGB4+ mitotic cells, whose offspring further segregated into a TNFRSF12Ahi subfraction that comprised about ten per cent of KRT5+ basal cells. This subpopulation formed clusters within terminal bronchioles and exhibited enriched clonogenic organoid growth activity. We created distal lung organoids with apical-out polarity to present ACE2 on the exposed external surface, facilitating infection of AT2 and basal cultures with SARS-CoV-2 and identifying club cells as a target population. This long-term, feeder-free culture of human distal lung organoids, coupled with single-cell analysis, identifies functional heterogeneity among basal cells and establishes a facile in vitro organoid model of human distal lung infections, including COVID-19-associated pneumonia.


Subject(s)
COVID-19/virology , Lung/cytology , Models, Biological , Organoids/cytology , Organoids/virology , SARS-CoV-2/physiology , Tissue Culture Techniques , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/metabolism , Alveolar Epithelial Cells/virology , COVID-19/metabolism , COVID-19/pathology , Cell Differentiation , Cell Division , Clone Cells/cytology , Clone Cells/metabolism , Clone Cells/virology , Humans , In Vitro Techniques , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/physiology , Integrin alpha6/analysis , Integrin beta4/analysis , Keratin-5/analysis , Organoids/metabolism , Pneumonia, Viral/metabolism , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , SARS-CoV-2/growth & development , Single-Cell Analysis , TWEAK Receptor/analysis
9.
bioRxiv ; 2020 Jul 27.
Article in English | MEDLINE | ID: mdl-32743583

ABSTRACT

The distal lung contains terminal bronchioles and alveoli that facilitate gas exchange and is affected by disorders including interstitial lung disease, cancer, and SARS-CoV-2-associated COVID-19 pneumonia. Investigations of these localized pathologies have been hindered by a lack of 3D in vitro human distal lung culture systems. Further, human distal lung stem cell identification has been impaired by quiescence, anatomic divergence from mouse and lack of lineage tracing and clonogenic culture. Here, we developed robust feeder-free, chemically-defined culture of distal human lung progenitors as organoids derived clonally from single adult human alveolar epithelial type II (AT2) or KRT5 + basal cells. AT2 organoids exhibited AT1 transdifferentiation potential, while basal cell organoids progressively developed lumens lined by differentiated club and ciliated cells. Organoids consisting solely of club cells were not observed. Upon single cell RNA-sequencing (scRNA-seq), alveolar organoids were composed of proliferative AT2 cells; however, basal organoid KRT5 + cells contained a distinct ITGA6 + ITGB4 + mitotic population whose proliferation segregated to a TNFRSF12A hi subfraction. Clonogenic organoid growth was markedly enriched within the TNFRSF12A hi subset of FACS-purified ITGA6 + ITGB4 + basal cells from human lung or derivative organoids. In vivo, TNFRSF12A + cells comprised ~10% of KRT5 + basal cells and resided in clusters within terminal bronchioles. To model COVID-19 distal lung disease, we everted the polarity of basal and alveolar organoids to rapidly relocate differentiated club and ciliated cells from the organoid lumen to the exterior surface, thus displaying the SARS-CoV-2 receptor ACE2 on the outwardly-facing apical aspect. Accordingly, basal and AT2 apical-out organoids were infected by SARS-CoV-2, identifying club cells as a novel target population. This long-term, feeder-free organoid culture of human distal lung alveolar and basal stem cells, coupled with single cell analysis, identifies unsuspected basal cell functional heterogeneity and exemplifies progenitor identification within a slowly proliferating human tissue. Further, our studies establish a facile in vitro organoid model for human distal lung infectious diseases including COVID-19-associated pneumonia.

10.
Nat Commun ; 10(1): 5587, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31811131

ABSTRACT

Elucidating the spectrum of epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) states in clinical samples promises insights on cancer progression and drug resistance. Using mass cytometry time-course analysis, we resolve lung cancer EMT states through TGFß-treatment and identify, through TGFß-withdrawal, a distinct MET state. We demonstrate significant differences between EMT and MET trajectories using a computational tool (TRACER) for reconstructing trajectories between cell states. In addition, we construct a lung cancer reference map of EMT and MET states referred to as the EMT-MET PHENOtypic STAte MaP (PHENOSTAMP). Using a neural net algorithm, we project clinical samples onto the EMT-MET PHENOSTAMP to characterize their phenotypic profile with single-cell resolution in terms of our in vitro EMT-MET analysis. In summary, we provide a framework to phenotypically characterize clinical samples in the context of in vitro EMT-MET findings which could help assess clinical relevance of EMT in cancer in future studies.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Lung Neoplasms/pathology , Algorithms , Cell Line, Tumor , Computational Biology , Cytophotometry/methods , Epithelial Cells/pathology , Humans , Lung Neoplasms/diagnostic imaging , Phenotype , Systems Biology , Transforming Growth Factor beta/metabolism
11.
Proc Natl Acad Sci U S A ; 115(18): E4294-E4303, 2018 05 01.
Article in English | MEDLINE | ID: mdl-29654148

ABSTRACT

An individual malignant tumor is composed of a heterogeneous collection of single cells with distinct molecular and phenotypic features, a phenomenon termed intratumoral heterogeneity. Intratumoral heterogeneity poses challenges for cancer treatment, motivating the need for combination therapies. Single-cell technologies are now available to guide effective drug combinations by accounting for intratumoral heterogeneity through the analysis of the signaling perturbations of an individual tumor sample screened by a drug panel. In particular, Mass Cytometry Time-of-Flight (CyTOF) is a high-throughput single-cell technology that enables the simultaneous measurements of multiple ([Formula: see text]40) intracellular and surface markers at the level of single cells for hundreds of thousands of cells in a sample. We developed a computational framework, entitled Drug Nested Effects Models (DRUG-NEM), to analyze CyTOF single-drug perturbation data for the purpose of individualizing drug combinations. DRUG-NEM optimizes drug combinations by choosing the minimum number of drugs that produce the maximal desired intracellular effects based on nested effects modeling. We demonstrate the performance of DRUG-NEM using single-cell drug perturbation data from tumor cell lines and primary leukemia samples.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Biomarkers, Tumor/metabolism , Computer Simulation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism , HeLa Cells , Humans
12.
Cell Stem Cell ; 21(1): 78-90.e6, 2017 07 06.
Article in English | MEDLINE | ID: mdl-28686870

ABSTRACT

Several cell populations have been reported to possess intestinal stem cell (ISC) activity during homeostasis and injury-induced regeneration. Here, we explored inter-relationships between putative mouse ISC populations by comparative RNA-sequencing (RNA-seq). The transcriptomes of multiple cycling ISC populations closely resembled Lgr5+ ISCs, the most well-defined ISC pool, but Bmi1-GFP+ cells were distinct and enriched for enteroendocrine (EE) markers, including Prox1. Prox1-GFP+ cells exhibited sustained clonogenic growth in vitro, and lineage-tracing of Prox1+ cells revealed long-lived clones during homeostasis and after radiation-induced injury in vivo. Single-cell mRNA-seq revealed two subsets of Prox1-GFP+ cells, one of which resembled mature EE cells while the other displayed low-level EE gene expression but co-expressed tuft cell markers, Lgr5 and Ascl2, reminiscent of label-retaining secretory progenitors. Our data suggest that the EE lineage, including mature EE cells, comprises a reservoir of homeostatic and injury-inducible ISCs, extending our understanding of cellular plasticity and stemness.


Subject(s)
Antigens, Differentiation/metabolism , Enteroendocrine Cells/metabolism , Intestinal Mucosa/injuries , Intestinal Mucosa/metabolism , Jejunum/injuries , Jejunum/metabolism , Stem Cells/metabolism , Animals , Antigens, Differentiation/genetics , Enteroendocrine Cells/pathology , Gene Expression Regulation , Intestinal Mucosa/pathology , Jejunum/pathology , Mice , Mice, Transgenic , Stem Cells/pathology
13.
Nat Protoc ; 11(7): 1264-79, 2016 07.
Article in English | MEDLINE | ID: mdl-27310265

ABSTRACT

High-throughput single-cell technologies provide an unprecedented view into cellular heterogeneity, yet they pose new challenges in data analysis and interpretation. In this protocol, we describe the use of Spanning-tree Progression Analysis of Density-normalized Events (SPADE), a density-based algorithm for visualizing single-cell data and enabling cellular hierarchy inference among subpopulations of similar cells. It was initially developed for flow and mass cytometry single-cell data. We describe SPADE's implementation and application using an open-source R package that runs on Mac OS X, Linux and Windows systems. A typical SPADE analysis on a 2.27-GHz processor laptop takes ∼5 min. We demonstrate the applicability of SPADE to single-cell RNA-seq data. We compare SPADE with recently developed single-cell visualization approaches based on the t-distribution stochastic neighborhood embedding (t-SNE) algorithm. We contrast the implementation and outputs of these methods for normal and malignant hematopoietic cells analyzed by mass cytometry and provide recommendations for appropriate use. Finally, we provide an integrative strategy that combines the strengths of t-SNE and SPADE to infer cellular hierarchy from high-dimensional single-cell data.


Subject(s)
Algorithms , Mass Spectrometry/methods , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Software , Animals , Antigens, CD/analysis , Hematologic Neoplasms/chemistry , Hematologic Neoplasms/pathology , Hematopoietic Stem Cells/chemistry , Hematopoietic Stem Cells/pathology , High-Throughput Screening Assays/methods , Humans , Mice , Stochastic Processes
14.
Aging (Albany NY) ; 7(9): 613-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26412380

ABSTRACT

We have found evidence suggesting that ARF and p53 are essential for tumor regression upon MYC inactivation through distinct mechanisms ARF through p53-independent affect, is required to for MYC to regulate the expression of genes that are required for both the induction of cellular senescence as well as recruitment of innate immune activation. Our observations have possible implications for mechanisms of therapeutic resistance to targeted oncogene inactivation.


Subject(s)
ADP-Ribosylation Factor 1/genetics , Aging/genetics , Immunity, Innate/genetics , Animals , Genes, p53 , Humans , Oncogenes/genetics
15.
Oncotarget ; 6(6): 3563-77, 2015 Feb 28.
Article in English | MEDLINE | ID: mdl-25784651

ABSTRACT

MYC-induced T-ALL exhibit oncogene addiction. Addiction to MYC is a consequence of both cell-autonomous mechanisms, such as proliferative arrest, cellular senescence, and apoptosis, as well as non-cell autonomous mechanisms, such as shutdown of angiogenesis, and recruitment of immune effectors. Here, we show, using transgenic mouse models of MYC-induced T-ALL, that the loss of either p19ARF or p53 abrogates the ability of MYC inactivation to induce sustained tumor regression. Loss of p53 or p19ARF, influenced the ability of MYC inactivation to elicit the shutdown of angiogenesis; however the loss of p19ARF, but not p53, impeded cellular senescence, as measured by SA-beta-galactosidase staining, increased expression of p16INK4A, and specific histone modifications. Moreover, comparative gene expression analysis suggested that a multitude of genes involved in the innate immune response were expressed in p19ARF wild-type, but not null, tumors upon MYC inactivation. Indeed, the loss of p19ARF, but not p53, impeded the in situ recruitment of macrophages to the tumor microenvironment. Finally, p19ARF null-associated gene signature prognosticated relapse-free survival in human patients with ALL. Therefore, p19ARF appears to be important to regulating cellular senescence and innate immune response that may contribute to the therapeutic response of ALL.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/immunology , Genes, myc , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , Animals , Cellular Senescence/genetics , Cellular Senescence/immunology , Disease Models, Animal , Gene Silencing , Humans , Immunity, Innate , Mice , Mice, Knockout , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/immunology
16.
PLoS Comput Biol ; 10(7): e1003664, 2014 Jul.
Article in English | MEDLINE | ID: mdl-25078380

ABSTRACT

A model-based gating strategy is developed for sorting cells and analyzing populations of single cells. The strategy, named CCAST, for Clustering, Classification and Sorting Tree, identifies a gating strategy for isolating homogeneous subpopulations from a heterogeneous population of single cells using a data-derived decision tree representation that can be applied to cell sorting. Because CCAST does not rely on expert knowledge, it removes human bias and variability when determining the gating strategy. It combines any clustering algorithm with silhouette measures to identify underlying homogeneous subpopulations, then applies recursive partitioning techniques to generate a decision tree that defines the gating strategy. CCAST produces an optimal strategy for cell sorting by automating the selection of gating markers, the corresponding gating thresholds and gating sequence; all of these parameters are typically manually defined. Even though CCAST is optimized for cell sorting, it can be applied for the identification and analysis of homogeneous subpopulations among heterogeneous single cell data. We apply CCAST on single cell data from both breast cancer cell lines and normal human bone marrow. On the SUM159 breast cancer cell line data, CCAST indicates at least five distinct cell states based on two surface markers (CD24 and EPCAM) and provides a gating sorting strategy that produces more homogeneous subpopulations than previously reported. When applied to normal bone marrow data, CCAST reveals an efficient strategy for gating T-cells without prior knowledge of the major T-cell subtypes and the markers that best define them. On the normal bone marrow data, CCAST also reveals two major mature B-cell subtypes, namely CD123+ and CD123- cells, which were not revealed by manual gating but show distinct intracellular signaling responses. More generally, the CCAST framework could be used on other biological and non-biological high dimensional data types that are mixtures of unknown homogeneous subpopulations.


Subject(s)
Cell Separation/methods , Computational Biology/methods , Computer Simulation , Algorithms , Biomarkers/chemistry , Cell Line, Tumor , Cluster Analysis , Humans
17.
Bioinformatics ; 30(3): 414-9, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24292937

ABSTRACT

MOTIVATION: For biological pathways, it is common to measure a gene expression time series after various knockdowns of genes that are putatively involved in the process of interest. These interventional time-resolved data are most suitable for the elucidation of dynamic causal relationships in signaling networks. Even with this kind of data it is still a major and largely unsolved challenge to infer the topology and interaction logic of the underlying regulatory network. RESULTS: In this work, we present a novel model-based approach involving Boolean networks to reconstruct small to medium-sized regulatory networks. In particular, we solve the problem of exact likelihood computation in Boolean networks with probabilistic exponential time delays. Simulations demonstrate the high accuracy of our approach. We apply our method to data of Ivanova et al. (2006), where RNA interference knockdown experiments were used to build a network of the key regulatory genes governing mouse stem cell maintenance and differentiation. In contrast to previous analyses of that data set, our method can identify feedback loops and provides new insights into the interplay of some master regulators in embryonic stem cell development. AVAILABILITY AND IMPLEMENTATION: The algorithm is implemented in the statistical language R. Code and documentation are available at Bioinformatics online. CONTACT: duemcke@mpipz.mpg.de or tresch@mpipz.mpg.de SUPPLEMENTARY INFORMATION: Supplementary Materials are available at Bioinfomatics online.


Subject(s)
Algorithms , Feedback, Physiological , Signal Transduction , Animals , Cell Differentiation , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression , Mice , Probability , RNA Interference
18.
Proc Natl Acad Sci U S A ; 106(16): 6447-52, 2009 Apr 21.
Article in English | MEDLINE | ID: mdl-19329492

ABSTRACT

Cellular decision making in differentiation, proliferation, or cell death is mediated by molecular signaling processes, which control the regulation and expression of genes. Vice versa, the expression of genes can trigger the activity of signaling pathways. We introduce and describe a statistical method called Dynamic Nested Effects Model (D-NEM) for analyzing the temporal interplay of cell signaling and gene expression. D-NEMs are Bayesian models of signal propagation in a network. They decompose observed time delays of multiple step signaling processes into single steps. Time delays are assumed to be exponentially distributed. Rate constants of signal propagation are model parameters, whose joint posterior distribution is assessed via Gibbs sampling. They hold information on the interplay of different forms of biological signal propagation. Molecular signaling in the cytoplasm acts at high rates, direct signal propagation via transcription and translation act at intermediate rates, while secondary effects operate at low rates. D-NEMs allow the dissection of biological processes into signaling and expression events, and analysis of cellular signal flow. An application of D-NEMs to embryonic stem cell development in mice reveals a feed-forward loop dominated network, which stabilizes the differentiated state of cells and points to Nanog as the key sensitizer of stem cells for differentiation stimuli.


Subject(s)
Gene Expression Regulation , Models, Genetic , Signal Transduction/genetics , Algorithms , Animals , Mice , Stem Cells/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...