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.
Genomics Proteomics Bioinformatics ; 19(6): 973-985, 2021 12.
Article in English | MEDLINE | ID: mdl-33581336

ABSTRACT

Continual reduction in sequencing cost is expanding the accessibility of genome sequencing data for routine clinical applications. However, the lack of methods to construct machine learning-based predictive models using these datasets has become a crucial bottleneck for the application of sequencing technology in clinics. Here, we develop a new algorithm, eTumorMetastasis, which transforms tumor functional mutations into network-based profiles and identifies network operational gene (NOG) signatures. NOG signatures model the tipping point at which a tumor cell shifts from a state that doesn't favor recurrence to one that does. We show that NOG signatures derived from genomic mutations of tumor founding clones (i.e., the 'most recent common ancestor' of the cells within a tumor) significantly distinguish the recurred and non-recurred breast tumors as well as outperform the most popular genomic test (i.e., Oncotype DX). These results imply that mutations of the tumor founding clones are associated with tumor recurrence and can be used to predict clinical outcomes. As such, predictive tools could be used in clinics to guide treatment routes. Finally, the concepts underlying the eTumorMetastasis pave the way for the application of genome sequencing in predictions for other complex genetic diseases. eTumorMetastasis pseudocode and related data used in this study are available at https://github.com/WangEdwinLab/eTumorMetastasis.


Subject(s)
Breast Neoplasms , Algorithms , Breast Neoplasms/genetics , Female , Genome , Humans , Machine Learning , Exome Sequencing
2.
Atherosclerosis ; 246: 78-86, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26761771

ABSTRACT

BACKGROUND: Strategies to reduce LDL-cholesterol involve reductions in cholesterol synthesis or absorption. We identified a familial hypercholesterolemia patient with an exceptional response to the cholesterol absorption inhibitor, ezetimibe. Niemann-Pick C 1-like 1 (NPC1L1) is the molecular target of ezetimibe. METHODS AND RESULTS: Sequencing identified nucleotide changes predicted to change amino acids 52 (L52P), 300 (I300T) and 489 (S489G) in exceptional NPC1L1. In silico analyses identified increased stability and cholesterol binding affinity in L52P-NPC1L1 versus WT-NPC1L1. HEK293 cells overexpressing WT-NPC1L1 or NPC1L1 harboring amino acid changes singly or in combination (Comb-NPC1L1) had reduced cholesterol uptake in Comb-NPC1L1 when ezetimibe was present. Cholesterol uptake was reduced by ezetimibe in L52P-NPC1L1, I300T-NPC1L1, but increased in S489G-NPC1L1 overexpressing cells. Immunolocalization studies found preferential plasma membrane localization of mutant NPC1L1 independent of ezetimibe. Flotillin 1 and 2 expression was reduced and binding to Comb-NPC1L1 was reduced independent of ezetimibe exposure. Proteomic analyses identified increased association with proteins that modulate intermediate filament proteins in Comb-NPC1L1 versus WT-NPC1L1 treated with ezetimibe. CONCLUSION: This is the first detailed analysis of the role of NPC1L1 mutations in an exceptional responder to ezetimibe. The results point to a complex set of events in which the combined mutations were shown to affect cholesterol uptake in the presence of ezetimibe. Proteomic analysis suggests that the exceptional response may also lie in the nature of interactions with cytosolic proteins.


Subject(s)
Anticholesteremic Agents/therapeutic use , Cholesterol, LDL/blood , Ezetimibe/therapeutic use , Hyperlipoproteinemia Type II/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mutation , Biomarkers/blood , DNA Mutational Analysis , Down-Regulation , Female , Genetic Markers , Genotype , HEK293 Cells , Humans , Hyperlipoproteinemia Type II/blood , Hyperlipoproteinemia Type II/drug therapy , Male , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Membrane Transport Proteins , Models, Molecular , Molecular Dynamics Simulation , Phenotype , Protein Binding , Protein Conformation , Proteomics/methods , Transfection , Treatment Outcome
3.
JAMA Oncol ; 2(1): 37-45, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26502222

ABSTRACT

IMPORTANCE: Decisions regarding adjuvant therapy in patients with stage II colorectal cancer (CRC) have been among the most challenging and controversial in oncology over the past 20 years. OBJECTIVE: To develop robust combinatory cancer hallmark-based gene signature sets (CSS sets) that more accurately predict prognosis and identify a subset of patients with stage II CRC who could gain survival benefits from adjuvant chemotherapy. DESIGN, SETTING, AND PARTICIPANTS: Thirteen retrospective studies of patients with stage II CRC who had clinical follow-up and adjuvant chemotherapy were analyzed. Respective totals of 162 and 843 patients from 2 and 11 independent cohorts were used as the discovery and validation cohorts, respectively. A total of 1005 patients with stage II CRC were included in the 13 cohorts. Among them, 84 of 416 patients in 3 independent cohorts received fluorouracil-based adjuvant chemotherapy. MAIN OUTCOMES AND MEASURES: Identification of CSS sets to predict relapse-free survival and identify a subset of patients with stage II CRC who could gain substantial survival benefits from fluorouracil-based adjuvant chemotherapy. RESULTS: Eight cancer hallmark-based gene signatures (30 genes each) were identified and used to construct CSS sets for determining prognosis. The CSS sets were validated in 11 independent cohorts of 767 patients with stage II CRC who did not receive adjuvant chemotherapy. The CSS sets accurately stratified patients into low-, intermediate-, and high-risk groups. Five-year relapse-free survival rates were 94%, 78%, and 45%, respectively, representing 60%, 28%, and 12% of patients with stage II disease. The 416 patients with CSS set-defined high-risk stage II CRC who received fluorouracil-based adjuvant chemotherapy showed a substantial gain in survival benefits from the treatment (ie, recurrence reduced by 30%-40% in 5 years). CONCLUSIONS AND RELEVANCE: The CSS sets substantially outperformed other prognostic predictors of stage 2 CRC. They are more accurate and robust for prognostic predictions and facilitate the identification of patients with stage II disease who could gain survival benefit from fluorouracil-based adjuvant chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Decision Support Techniques , Gene Expression Profiling , Neoplasm Recurrence, Local , Antimetabolites, Antineoplastic/therapeutic use , Chemotherapy, Adjuvant , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease-Free Survival , Fluorouracil/therapeutic use , Gene Expression Profiling/methods , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Neoplasm Staging , Oligonucleotide Array Sequence Analysis , Patient Selection , Phenotype , Precision Medicine , Predictive Value of Tests , Reproducibility of Results , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors , Treatment Outcome
4.
Semin Cancer Biol ; 30: 4-12, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24747696

ABSTRACT

Tumor genome sequencing leads to documenting thousands of DNA mutations and other genomic alterations. At present, these data cannot be analyzed adequately to aid in the understanding of tumorigenesis and its evolution. Moreover, we have little insight into how to use these data to predict clinical phenotypes and tumor progression to better design patient treatment. To meet these challenges, we discuss a cancer hallmark network framework for modeling genome sequencing data to predict cancer clonal evolution and associated clinical phenotypes. The framework includes: (1) cancer hallmarks that can be represented by a few molecular/signaling networks. 'Network operational signatures' which represent gene regulatory logics/strengths enable to quantify state transitions and measures of hallmark traits. Thus, sets of genomic alterations which are associated with network operational signatures could be linked to the state/measure of hallmark traits. The network operational signature transforms genotypic data (i.e., genomic alterations) to regulatory phenotypic profiles (i.e., regulatory logics/strengths), to cellular phenotypic profiles (i.e., hallmark traits) which lead to clinical phenotypic profiles (i.e., a collection of hallmark traits). Furthermore, the framework considers regulatory logics of the hallmark networks under tumor evolutionary dynamics and therefore also includes: (2) a self-promoting positive feedback loop that is dominated by a genomic instability network and a cell survival/proliferation network is the main driver of tumor clonal evolution. Surrounding tumor stroma and its host immune systems shape the evolutionary paths; (3) cell motility initiating metastasis is a byproduct of the above self-promoting loop activity during tumorigenesis; (4) an emerging hallmark network which triggers genome duplication dominates a feed-forward loop which in turn could act as a rate-limiting step for tumor formation; (5) mutations and other genomic alterations have specific patterns and tissue-specificity, which are driven by aging and other cancer-inducing agents. This framework represents the logics of complex cancer biology as a myriad of phenotypic complexities governed by a limited set of underlying organizing principles. It therefore adds to our understanding of tumor evolution and tumorigenesis, and moreover, potential usefulness of predicting tumors' evolutionary paths and clinical phenotypes. Strategies of using this framework in conjunction with genome sequencing data in an attempt to predict personalized drug targets, drug resistance, and metastasis for cancer patients, as well as cancer risks for healthy individuals are discussed. Accurate prediction of cancer clonal evolution and clinical phenotypes will have substantial impact on timely diagnosis, personalized treatment and personalized prevention of cancer.


Subject(s)
Gene Regulatory Networks/genetics , Genomics/methods , Models, Genetic , Neoplasms/genetics , Precision Medicine/methods , Genome, Human , Humans , Phenotype
5.
PLoS One ; 9(11): e113190, 2014.
Article in English | MEDLINE | ID: mdl-25409505

ABSTRACT

The androgen receptor (AR) remains an important contributor to the neoplastic evolution of prostate cancer (CaP). CaP progression is linked to several somatic AR mutational changes that endow upon the AR dramatic gain-of-function properties. One of the most common somatic mutations identified is Thr877-to-Ala (T877A), located in the ligand-binding domain, that results in a receptor capable of promiscuous binding and activation by a variety of steroid hormones and ligands including estrogens, progestins, glucocorticoids, and several anti-androgens. In an attempt to further define somatic mutated AR gain-of-function properties, as a consequence of its promiscuous ligand binding, we undertook a proteomic/network analysis approach to characterize the protein interactome of the mutant T877A-AR in LNCaP cells under eight different ligand-specific treatments (dihydrotestosterone, mibolerone, R1881, testosterone, estradiol, progesterone, dexamethasone, and cyproterone acetate). In extending the analysis of our multi-ligand complexes of the mutant T877A-AR we observed significant enrichment of specific complexes between normal and primary prostatic tumors, which were furthermore correlated with known clinical outcomes. Further analysis of certain mutant T877A-AR complexes showed specific population preferences distinguishing primary prostatic disease between white (non-Hispanic) vs. African-American males. Moreover, these cancer-related AR-protein complexes demonstrated predictive survival outcomes specific to CaP, and not for breast, lung, lymphoma or medulloblastoma cancers. Our study, by coupling data generated by our proteomics to network analysis of clinical samples, has helped to define real and novel biological pathways in complicated gain-of-function AR complex systems.


Subject(s)
Black or African American/genetics , Gonadal Steroid Hormones/pharmacology , Prostatic Neoplasms/ethnology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , White People/genetics , Binding Sites , Cell Line, Tumor , Gonadal Steroid Hormones/chemistry , Humans , Male , Models, Molecular , Prognosis , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Structure, Secondary , Proteomics , Receptors, Androgen/chemistry , Survival Analysis , White People/ethnology
6.
Cell Rep ; 5(1): 216-23, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24075989

ABSTRACT

Individual cancer cells carry a bewildering number of distinct genomic alterations (e.g., copy number variations and mutations), making it a challenge to uncover genomic-driven mechanisms governing tumorigenesis. Here, we performed exome sequencing on several breast cancer cell lines that represent two subtypes, luminal and basal. We integrated these sequencing data and functional RNAi screening data (for the identification of genes that are essential for cell proliferation and survival) onto a human signaling network. Two subtype-specific networks that potentially represent core-signaling mechanisms underlying tumorigenesis were identified. Within both networks, we found that genes were differentially affected in different cell lines; i.e., in some cell lines a gene was identified through RNAi screening, whereas in others it was genomically altered. Interestingly, we found that highly connected network genes could be used to correctly classify breast tumors into subtypes on the basis of genomic alterations. Further, the networks effectively predicted subtype-specific drug targets, which were experimentally validated.


Subject(s)
Breast Neoplasms/genetics , DNA Copy Number Variations , Mutation , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Exome , Female , Humans , Molecular Targeted Therapy , Signal Transduction
7.
Semin Cancer Biol ; 23(4): 279-85, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23791722

ABSTRACT

Recent tumor genome sequencing confirmed that one tumor often consists of multiple cell subpopulations (clones) which bear different, but related, genetic profiles such as mutation and copy number variation profiles. Thus far, one tumor has been viewed as a whole entity in cancer functional studies. With the advances of genome sequencing and computational analysis, we are able to quantify and computationally dissect clones from tumors, and then conduct clone-based analysis. Emerging technologies such as single-cell genome sequencing and RNA-Seq could profile tumor clones. Thus, we should reconsider how to conduct cancer systems biology studies in the genome sequencing era. We will outline new directions for conducting cancer systems biology by considering that genome sequencing technology can be used for dissecting, quantifying and genetically characterizing clones from tumors. Topics discussed in Part 1 of this review include computationally quantifying of tumor subpopulations; clone-based network modeling, cancer hallmark-based networks and their high-order rewiring principles and the principles of cell survival networks of fast-growing clones.


Subject(s)
Genome, Human/genetics , Neoplasms/genetics , Sequence Analysis, DNA/methods , Systems Biology/methods , Apoptosis/genetics , Cell Cycle/genetics , Gene Regulatory Networks , Humans , Models, Genetic , Neoplasms/pathology , Single-Cell Analysis/methods
8.
Semin Cancer Biol ; 23(4): 286-92, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23792107

ABSTRACT

A tumor often consists of multiple cell subpopulations (clones). Current chemo-treatments often target one clone of a tumor. Although the drug kills that clone, other clones overtake it and the tumor recurs. Genome sequencing and computational analysis allows to computational dissection of clones from tumors, while singe-cell genome sequencing including RNA-Seq allows profiling of these clones. This opens a new window for treating a tumor as a system in which clones are evolving. Future cancer systems biology studies should consider a tumor as an evolving system with multiple clones. Therefore, topics discussed in Part 2 of this review include evolutionary dynamics of clonal networks, early-warning signals (e.g., genome duplication events) for formation of fast-growing clones, dissecting tumor heterogeneity, and modeling of clone-clone-stroma interactions for drug resistance. The ultimate goal of the future systems biology analysis is to obtain a 'whole-system' understanding of a tumor and therefore provides a more efficient and personalized management strategies for cancer patients.


Subject(s)
Genome, Human/genetics , Neoplasms/genetics , Sequence Analysis, DNA/methods , Systems Biology/methods , Cell Lineage/genetics , Gene Regulatory Networks , Humans , Models, Genetic , Neoplasms/pathology , Single-Cell Analysis/methods , Tumor Microenvironment/genetics
9.
Integr Biol (Camb) ; 3(10): 1020-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21901193

ABSTRACT

The androgen receptor (AR) is a ligand-inducible transcription factor, a member of the nuclear receptor superfamily, which plays an important role in the development and progression of prostate cancer (CaP). The transformation to CaP has been linked to several somatic AR gene mutations and changes in AR protein complex formation, which in turn increase the potential activity of the receptor. Thus, to address the mechanism of AR-mediated neoplastic transformation, we developed in vitro methodology to isolate and characterize, via mass spectrometry, AR complexes of three AR genetic variants: wild type-AR, and two somatic gain-of-function AR prostatic mutants (T877A-AR and 0CAG-AR isoforms). To fully characterize the significance of our large raw data set, we employed a sophisticated systems biology approach to create an integrative protein-interaction network profile for each AR isoform. Our comparative analysis identified subnetwork cluster profiles for AR isoforms (WT, T877A, and 0CAG) that segregated AR isoforms on the basis of androgen stimulation conditions and mutant aggressiveness. Furthermore, results from additional correlative gene microarray analysis studies of all three AR isoform (WT, T877A, 0CAG) subnetwork clusters were assessed and found to be significantly enriched in tumor versus normal prostate tissues. We also identified two AR-interaction clusters, containing 21 and 30 proteins, respectively, that showed unfavourable prognosis outcome of recurrent cancers, on the basis of PSA, Gleason score and combined PSA/Gleason score. In conclusion, we have characterized a large panel of novel AR-interacting proteins, through a combined proteomics/systems biology screen, that are of clinical relevance and could potentially serve as novel markers for diagnosis and prognosis of CaP.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Animals , COS Cells , Cell Transformation, Neoplastic/genetics , Chlorocebus aethiops , Chromatography, Liquid , Disease Progression , Genetic Variation , Humans , Male , Polymorphism, Single Nucleotide , Prostatic Neoplasms/genetics , Protein Interaction Maps , Protein Isoforms , Proteomics , Receptors, Androgen/genetics , Systems Biology/methods , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...