Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 8(1): 16142, 2018 Oct 26.
Article in English | MEDLINE | ID: mdl-30367081

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has been fixed in the paper.

2.
Sci Rep ; 8(1): 13658, 2018 09 12.
Article in English | MEDLINE | ID: mdl-30209281

ABSTRACT

Quantitative analysis of morphological changes in a cell nucleus is important for the understanding of nuclear architecture and its relationship with pathological conditions such as cancer. However, dimensionality of imaging data, together with a great variability of nuclear shapes, presents challenges for 3D morphological analysis. Thus, there is a compelling need for robust 3D nuclear morphometric techniques to carry out population-wide analysis. We propose a new approach that combines modeling, analysis, and interpretation of morphometric characteristics of cell nuclei and nucleoli in 3D. We used robust surface reconstruction that allows accurate approximation of 3D object boundary. Then, we computed geometric morphological measures characterizing the form of cell nuclei and nucleoli. Using these features, we compared over 450 nuclei with about 1,000 nucleoli of epithelial and mesenchymal prostate cancer cells, as well as 1,000 nuclei with over 2,000 nucleoli from serum-starved and proliferating fibroblast cells. Classification of sets of 9 and 15 cells achieved accuracy of 95.4% and 98%, respectively, for prostate cancer cells, and 95% and 98% for fibroblast cells. To our knowledge, this is the first attempt to combine these methods for 3D nuclear shape modeling and morphometry into a highly parallel pipeline workflow for morphometric analysis of thousands of nuclei and nucleoli in 3D.


Subject(s)
Cell Nucleolus/physiology , Cell Nucleus/physiology , Epithelial Cells/physiology , Fibroblasts/physiology , Imaging, Three-Dimensional/methods , Prostatic Neoplasms/pathology , Cell Nucleolus/pathology , Cell Nucleus/pathology , Humans , Male , Tumor Cells, Cultured
3.
Pharmacogenomics ; 19(7): 629-650, 2018 05.
Article in English | MEDLINE | ID: mdl-29697304

ABSTRACT

This Perspective provides examples of current and future applications of deep learning in pharmacogenomics, including: identification of novel regulatory variants located in noncoding domains of the genome and their function as applied to pharmacoepigenomics; patient stratification from medical records; and the mechanistic prediction of drug response, targets and their interactions. Deep learning encapsulates a family of machine learning algorithms that has transformed many important subfields of artificial intelligence over the last decade, and has demonstrated breakthrough performance improvements on a wide range of tasks in biomedicine. We anticipate that in the future, deep learning will be widely used to predict personalized drug response and optimize medication selection and dosing, using knowledge extracted from large and complex molecular, epidemiological, clinical and demographic datasets.


Subject(s)
Deep Learning , Models, Educational , Pharmacogenetics/education , Pharmacogenetics/trends , Algorithms , Databases as Topic , Deep Learning/trends , Humans , Neural Networks, Computer
4.
Pharmacogenomics ; 19(5): 413-434, 2018 04.
Article in English | MEDLINE | ID: mdl-29400612

ABSTRACT

AIM: 'Pharmacoepigenomics' methods informed by omics datasets and pre-existing knowledge have yielded discoveries in neuropsychiatric pharmacogenomics. Now we evaluate the generality of these methods by discovering an extended warfarin pharmacogenomics pathway. MATERIALS & METHODS: We developed the pharmacoepigenomics informatics pipeline, a scalable multi-omics variant screening pipeline for pharmacogenomics, and conducted an experiment in the genomics of warfarin. RESULTS: We discovered known and novel pharmacogenomics variants and genes, both coding and regulatory, for warfarin response, including adverse events. Such genes and variants cluster in a warfarin response pathway consolidating known and novel warfarin response variants and genes. CONCLUSION: These results can inform a new warfarin test. The pharmacoepigenomics informatics pipeline may be able to discover new pharmacogenomics markers in other drug-disease systems.


Subject(s)
Anticoagulants/therapeutic use , Computational Biology , Pharmacogenetics , Warfarin/therapeutic use , Anticoagulants/adverse effects , Blood Coagulation Disorders/drug therapy , Blood Coagulation Disorders/genetics , Genetic Variation/genetics , Genome-Wide Association Study , Humans , Lithium Compounds/therapeutic use , Polymorphism, Single Nucleotide , Warfarin/adverse effects
5.
Methods ; 123: 102-118, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28385536

ABSTRACT

The pharmacoepigenome can be defined as the active, noncoding province of the genome including canonical spatial and temporal regulatory mechanisms of gene regulation that respond to xenobiotic stimuli. Many psychotropic drugs that have been in clinical use for decades have ill-defined mechanisms of action that are beginning to be resolved as we understand the transcriptional hierarchy and dynamics of the nucleus. In this review, we describe spatial, temporal and biomechanical mechanisms mediated by psychotropic medications. Focus is placed on a bioinformatics pipeline that can be used both for detection of pharmacoepigenomic variants that discretize drug response and adverse events to improve pharmacogenomic testing, and for the discovery of novel CNS therapeutics. This approach integrates the functional topology and dynamics of the transcriptional hierarchy of the pharmacoepigenome, gene variant-driven identification of pharmacogenomic regulatory domains, and mesoscale mapping for the discovery of novel CNS pharmacodynamic pathways in human brain. Examples of the application of this pipeline are provided, including the discovery of valproic acid (VPA) mediated transcriptional reprogramming of neuronal cell fate following injury, and mapping of a CNS pathway glutamatergic pathway for the mood stabilizer lithium. These examples in regulatory pharmacoepigenomics illustrate how ongoing research using the 4D nucleome provides a foundation to further insight into previously unrecognized psychotropic drug pharmacodynamic pathways in the human CNS.


Subject(s)
Computational Biology/methods , Genome, Human , Nerve Tissue Proteins/genetics , Pharmacogenetics/methods , Psychotropic Drugs/therapeutic use , Brain/drug effects , Brain/metabolism , Brain/physiopathology , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Nucleus/ultrastructure , Chromosomes, Human/drug effects , Chromosomes, Human/metabolism , Chromosomes, Human/ultrastructure , Circadian Rhythm/physiology , Data Mining/methods , Gene Expression Regulation , Humans , Lithium/therapeutic use , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Transcription, Genetic , Valproic Acid/therapeutic use
6.
Pharm Res ; 34(8): 1658-1672, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28271248

ABSTRACT

OBJECTIVES: To determine the mechanism of action of valproic acid (VPA) in the adult central nervous system (CNS) following traumatic brain injury (TBI) and hemorrhagic shock (HS). METHODS: Data were analyzed from different sources, including experiments in a porcine model, data from postmortem human brain, published studies, public and commercial databases. RESULTS: The transcriptional program in the CNS following TBI, HS, and VPA treatment includes activation of regulatory pathways that enhance neurogenesis and suppress gliogenesis. Genes which encode the transcription factors (TFs) that specify neuronal cell fate, including MEF2D, MYT1L, NEUROD1, PAX6 and TBR1, and their target genes, are induced by VPA. VPA represses genes responsible for oligodendrogenesis, maintenance of white matter, T-cell activation, angiogenesis, and endothelial cell proliferation, adhesion and chemotaxis. NEUROD1 has regulatory interactions with 38% of the genes regulated by VPA in a swine model of TBI and HS in adult brain. Hi-C spatial mapping of a VPA pharmacogenomic SNP in the GRIN2B gene shows it is part of a transcriptional hub that contacts 12 genes that mediate chromatin-mediated neurogenesis and neuroplasticity. CONCLUSIONS: Following TBI and HS, this study shows that VPA administration acts in the adult brain through differential activation of TFs responsible for neurogenesis, genes responsible for neuroplasticity, and repression of TFs that specify oligodendrocyte cell fate, endothelial cell chemotaxis and angiogenesis. Short title: Mechanism of action of valproic acid in traumatic brain injury.


Subject(s)
Anticonvulsants/pharmacology , Brain Injuries, Traumatic/metabolism , Brain/drug effects , Gene Regulatory Networks , Shock, Hemorrhagic/metabolism , Transcription Factors/metabolism , Valproic Acid/pharmacology , Adult , Animals , Brain/metabolism , Brain/pathology , Brain Injuries, Traumatic/pathology , Cell Line, Tumor , Gene Expression , Humans , Neurogenesis/genetics , Neuronal Plasticity/genetics , Neurons/pathology , Oligodendroglia/pathology , Pharmacogenetics , Rodentia , Shock, Hemorrhagic/pathology , Swine , Transcription Factors/genetics , Transcriptional Activation
7.
Pharmacogenomics ; 16(14): 1565-83, 2015.
Article in English | MEDLINE | ID: mdl-26340055

ABSTRACT

AIM: To provide insight into potential regulatory mechanisms of gene expression underlying addiction, analgesia, psychotropic drug response and adverse drug events, genome-wide association studies searching for variants associated with these phenotypes has been undertaken with limited success. We undertook analysis of these results with the aim of applying epigenetic knowledge to aid variant discovery and interpretation. METHODS: We applied conditional imputation to results from 26 genome-wide association studies and three candidate gene-association studies. The analysis workflow included data from chromatin conformation capture, chromatin state annotation, DNase I hypersensitivity, hypomethylation, anatomical localization and biochronicity. We also made use of chromatin state data from the epigenome roadmap, transcription factor-binding data, spatial maps from published Hi-C datasets and 'guilt by association' methods. RESULTS: We identified 31 pharmacoepigenomic SNPs from a total of 2024 variants in linkage disequilibrium with lead SNPs, of which only 6% were coding variants. Interrogation of chromatin state using our workflow and the epigenome roadmap showed agreement on 34 of 35 tissue assignments to regulatory elements including enhancers and promoters. Loop boundary domains were inferred by association with CTCF (CCCTC-binding factor) and cohesin, suggesting proximity to topologically associating domain boundaries and enhancer clusters. Spatial interactions between enhancer-promoter pairs detected both known and previously unknown mechanisms. Addiction and analgesia SNPs were common in relevant populations and exhibited large effect sizes, whereas a SNP located in the promoter of the SLC1A2 gene exhibited a moderate effect size for lithium response in bipolar disorder in patients of European ancestry. SNPs associated with drug-induced organ injury were rare but exhibited the largest effect sizes, consistent with the published literature. CONCLUSION: This work demonstrates that an in silico bioinformatics-based approach using integrative analysis of a diversity of molecular and morphological data types can discover pharmacoepigenomic variants that are suitable candidates for further validation in cell lines, animal models and human clinical trials.


Subject(s)
Chromosome Mapping/methods , Epigenomics/methods , Psychotropic Drugs/pharmacology , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromosomal Proteins, Non-Histone/genetics , Computational Biology , Computer Simulation , DNA Methylation , Deoxyribonuclease I/genetics , Genetic Variation , Genome-Wide Association Study , Humans , Polymorphism, Single Nucleotide , Transcription Factors/genetics , White People , Cohesins
8.
Pharmacogenomics ; 16(14): 1547-63, 2015.
Article in English | MEDLINE | ID: mdl-26343379

ABSTRACT

AIM: A regulatory network in the human brain mediating lithium response in bipolar patients was revealed by analysis of functional SNPs from genome-wide association studies (GWAS) and published gene association studies, followed by epigenome mapping. METHODS: An initial set of 23,312 SNPs in linkage disequilibrium with lead SNPs, and sub-threshold GWAS SNPs rescued by pathway analysis, were studied in the same populations. These were assessed using our workflow and annotation by the epigenome roadmap consortium. RESULTS: Twenty-seven percent of 802 SNPs that were associated with lithium response (13 published studies gene association studies and two GWAS) were shared in common with 1281 SNPs from 18 GWAS examining psychiatric disorders and adverse events associated with lithium treatment. Nineteen SNPs were annotated as active regulatory elements such as enhancers and promoters in a tissue-specific manner. They were located within noncoding regions of ten genes: ANK3, ARNTL, CACNA1C, CACNG2, CDKN1A, CREB1, GRIA2, GSK3B, NR1D1 and SLC1A2. Following gene set enrichment and pathway analysis, these genes were found to be significantly associated (p = 10(-27); Fisher exact test) with an AMPA2 glutamate receptor network in human brain. Our workflow results showed concordance with annotation of regulatory elements from the epigenome roadmap. Analysis of cognate mRNA and enhancer RNA exhibited patterns consistent with an integrated pathway in human brain. CONCLUSION: This pharmacoepigenomic regulatory pathway is located in the same brain regions that exhibit tissue volume loss in bipolar disorder. Although in silico analysis requires biological validation, the approach provides value for identification of candidate variants that may be used in pharmacogenomic testing to identify bipolar patients likely to respond to lithium.


Subject(s)
Antimanic Agents/therapeutic use , Bipolar Disorder/drug therapy , Bipolar Disorder/genetics , Epigenesis, Genetic/genetics , Glutamic Acid/genetics , Glutamic Acid/metabolism , Lithium Compounds/therapeutic use , Chromosome Mapping , Computer Simulation , Genome-Wide Association Study , Humans , In Situ Hybridization , Polymorphism, Single Nucleotide , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, AMPA/genetics
9.
Pharmacogenomics ; 16(14): 1649-69, 2015.
Article in English | MEDLINE | ID: mdl-26338265

ABSTRACT

The 4D nucleome has the potential to render challenges in neuropsychiatric pharmacogenomics more tractable. The epigenome roadmap consortium has demonstrated the critical role that noncoding regions of the human genome play in determination of human phenotype. Chromosome conformation capture methods have revealed the 4D organization of the nucleus, bringing interactions between distant regulatory elements into close spatial proximity in a periodic manner. These functional interactions have the potential to elucidate mechanisms of CNS drug response and side effects that previously have been unrecognized. This perspective assesses recent advances likely to reveal novel pharmacodynamic regulatory pathways in human brain, charting a future new avenue of pharmacogenomics research, using the spatial and temporal architecture of the human epigenome as its foundation.


Subject(s)
Epigenesis, Genetic/genetics , Epigenomics/trends , Pharmacogenetics/trends , Psychotropic Drugs/pharmacology , Genome, Human , Humans , Mental Disorders/drug therapy , Mental Disorders/genetics , Psychotropic Drugs/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...