Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
Add more filters

Publication year range
1.
PLoS Med ; 17(11): e1003323, 2020 11.
Article in English | MEDLINE | ID: mdl-33147277

ABSTRACT

BACKGROUND: The tumor microenvironment (TME) is increasingly appreciated as an important determinant of cancer outcome, including in multiple myeloma (MM). However, most myeloma microenvironment studies have been based on bone marrow (BM) aspirates, which often do not fully reflect the cellular content of BM tissue itself. To address this limitation in myeloma research, we systematically characterized the whole bone marrow (WBM) microenvironment during premalignant, baseline, on treatment, and post-treatment phases. METHODS AND FINDINGS: Between 2004 and 2019, 998 BM samples were taken from 436 patients with newly diagnosed MM (NDMM) at the University of Arkansas for Medical Sciences in Little Rock, Arkansas, United States of America. These patients were 61% male and 39% female, 89% White, 8% Black, and 3% other/refused, with a mean age of 58 years. Using WBM and matched cluster of differentiation (CD)138-selected tumor gene expression to control for tumor burden, we identified a subgroup of patients with an adverse TME associated with 17 fewer months of progression-free survival (PFS) (95% confidence interval [CI] 5-29, 49-69 versus 70-82 months, χ2 p = 0.001) and 15 fewer months of overall survival (OS; 95% CI -1 to 31, 92-120 versus 113-129 months, χ2 p = 0.036). Using immunohistochemistry-validated computational tools that identify distinct cell types from bulk gene expression, we showed that the adverse outcome was correlated with elevated CD8+ T cell and reduced granulocytic cell proportions. This microenvironment develops during the progression of premalignant to malignant disease and becomes less prevalent after therapy, in which it is associated with improved outcomes. In patients with quantified International Staging System (ISS) stage and 70-gene Prognostic Risk Score (GEP-70) scores, taking the microenvironment into consideration would have identified an additional 40 out of 290 patients (14%, premutation p = 0.001) with significantly worse outcomes (PFS, 95% CI 6-36, 49-73 versus 74-90 months) who were not identified by existing clinical (ISS stage III) and tumor (GEP-70) criteria as high risk. The main limitations of this study are that it relies on computationally identified cell types and that patients were treated with thalidomide rather than current therapies. CONCLUSIONS: In this study, we observe that granulocyte signatures in the MM TME contribute to a more accurate prognosis. This implies that future researchers and clinicians treating patients should quantify TME components, in particular monocytes and granulocytes, which are often ignored in microenvironment studies.


Subject(s)
Bone Marrow/pathology , Multiple Myeloma/diagnosis , Multiple Myeloma/pathology , Tumor Microenvironment , Adult , Cohort Studies , Female , Humans , Male , Middle Aged , Multiple Myeloma/drug therapy , Prognosis , Tumor Burden
2.
Mol Syst Biol ; 13(3): 919, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28320772

ABSTRACT

Managing trade-offs through gene regulation is believed to confer resilience to a microbial community in a fluctuating resource environment. To investigate this hypothesis, we imposed a fluctuating environment that required the sulfate-reducer Desulfovibrio vulgaris to undergo repeated ecologically relevant shifts between retaining metabolic independence (active capacity for sulfate respiration) and becoming metabolically specialized to a mutualistic association with the hydrogen-consuming Methanococcus maripaludis Strikingly, the microbial community became progressively less proficient at restoring the environmentally relevant physiological state after each perturbation and most cultures collapsed within 3-7 shifts. Counterintuitively, the collapse phenomenon was prevented by a single regulatory mutation. We have characterized the mechanism for collapse by conducting RNA-seq analysis, proteomics, microcalorimetry, and single-cell transcriptome analysis. We demonstrate that the collapse was caused by conditional gene regulation, which drove precipitous decline in intracellular abundance of essential transcripts and proteins, imposing greater energetic burden of regulation to restore function in a fluctuating environment.


Subject(s)
Desulfovibrio vulgaris/growth & development , Methanococcus/growth & development , Systems Biology/methods , Desulfovibrio vulgaris/genetics , Directed Molecular Evolution , Gene Expression Profiling , Methanococcus/genetics , Oxidation-Reduction , Phenotype , Proteomics , Sequence Analysis, RNA , Single-Cell Analysis , Sulfates/metabolism
3.
Nucleic Acids Res ; 43(13): e87, 2015 Jul 27.
Article in English | MEDLINE | ID: mdl-25873626

ABSTRACT

The cMonkey integrated biclustering algorithm identifies conditionally co-regulated modules of genes (biclusters). cMonkey integrates various orthogonal pieces of information which support evidence of gene co-regulation, and optimizes biclusters to be supported simultaneously by one or more of these prior constraints. The algorithm served as the cornerstone for constructing the first global, predictive Environmental Gene Regulatory Influence Network (EGRIN) model for a free-living cell, and has now been applied to many more organisms. However, due to its computational inefficiencies, long run-time and complexity of various input data types, cMonkey was not readily usable by the wider community. To address these primary concerns, we have significantly updated the cMonkey algorithm and refactored its implementation, improving its usability and extendibility. These improvements provide a fully functioning and user-friendly platform for building co-regulated gene modules and the tools necessary for their exploration and interpretation. We show, via three separate analyses of data for E. coli, M. tuberculosis and H. sapiens, that the updated algorithm and inclusion of novel scoring functions for new data types (e.g. ChIP-seq and transcription factor over-expression [TFOE]) improve discovery of biologically informative co-regulated modules. The complete cMonkey2 software package, including source code, is available at https://github.com/baliga-lab/cmonkey2.


Subject(s)
Gene Expression Regulation , Software , Algorithms , Carcinoma, Squamous Cell/genetics , Chromatin Immunoprecipitation , Escherichia coli/genetics , Gene Expression Regulation, Bacterial , Lung Neoplasms/genetics , Mycobacterium tuberculosis/genetics , Regulon , Sequence Analysis, DNA , Transcription Factors/metabolism
4.
Genome Res ; 23(11): 1839-51, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24089473

ABSTRACT

Methanogens catalyze the critical methane-producing step (called methanogenesis) in the anaerobic decomposition of organic matter. Here, we present the first predictive model of global gene regulation of methanogenesis in a hydrogenotrophic methanogen, Methanococcus maripaludis. We generated a comprehensive list of genes (protein-coding and noncoding) for M. maripaludis through integrated analysis of the transcriptome structure and a newly constructed Peptide Atlas. The environment and gene-regulatory influence network (EGRIN) model of the strain was constructed from a compendium of transcriptome data that was collected over 58 different steady-state and time-course experiments that were performed in chemostats or batch cultures under a spectrum of environmental perturbations that modulated methanogenesis. Analyses of the EGRIN model have revealed novel components of methanogenesis that included at least three additional protein-coding genes of previously unknown function as well as one noncoding RNA. We discovered that at least five regulatory mechanisms act in a combinatorial scheme to intercoordinate key steps of methanogenesis with different processes such as motility, ATP biosynthesis, and carbon assimilation. Through a combination of genetic and environmental perturbation experiments we have validated the EGRIN-predicted role of two novel transcription factors in the regulation of phosphate-dependent repression of formate dehydrogenase-a key enzyme in the methanogenesis pathway. The EGRIN model demonstrates regulatory affiliations within methanogenesis as well as between methanogenesis and other cellular functions.


Subject(s)
Genes, Archaeal , Metabolic Networks and Pathways/genetics , Methane/biosynthesis , Methanococcus/enzymology , Methanococcus/genetics , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Formate Dehydrogenases/genetics , Gene Expression Profiling , Gene Expression Regulation, Archaeal , Gene-Environment Interaction , Hydrogen/metabolism , Methanococcus/metabolism , Models, Genetic , Sequence Deletion
5.
Nucleic Acids Res ; 42(Database issue): D184-90, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24271392

ABSTRACT

The ease of generating high-throughput data has enabled investigations into organismal complexity at the systems level through the inference of networks of interactions among the various cellular components (genes, RNAs, proteins and metabolites). The wider scientific community, however, currently has limited access to tools for network inference, visualization and analysis because these tasks often require advanced computational knowledge and expensive computing resources. We have designed the network portal (http://networks.systemsbiology.net) to serve as a modular database for the integration of user uploaded and public data, with inference algorithms and tools for the storage, visualization and analysis of biological networks. The portal is fully integrated into the Gaggle framework to seamlessly exchange data with desktop and web applications and to allow the user to create, save and modify workspaces, and it includes social networking capabilities for collaborative projects. While the current release of the database contains networks for 13 prokaryotic organisms from diverse phylogenetic clades (4678 co-regulated gene modules, 3466 regulators and 9291 cis-regulatory motifs), it will be rapidly populated with prokaryotic and eukaryotic organisms as relevant data become available in public repositories and through user input. The modular architecture, simple data formats and open API support community development of the portal.


Subject(s)
Databases, Genetic , Gene Regulatory Networks , Algorithms , Archaea/genetics , Archaea/metabolism , Bacteria/genetics , Bacteria/metabolism , Computer Graphics , Gene Expression Profiling , Internet , Nucleotide Motifs , Regulatory Elements, Transcriptional , Software , Systems Integration , Transcription Factors/metabolism
6.
Nucleic Acids Res ; 42(18): 11291-303, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25232098

ABSTRACT

The resilience of Mycobacterium tuberculosis (MTB) is largely due to its ability to effectively counteract and even take advantage of the hostile environments of a host. In order to accelerate the discovery and characterization of these adaptive mechanisms, we have mined a compendium of 2325 publicly available transcriptome profiles of MTB to decipher a predictive, systems-scale gene regulatory network model. The resulting modular organization of 98% of all MTB genes within this regulatory network was rigorously tested using two independently generated datasets: a genome-wide map of 7248 DNA-binding locations for 143 transcription factors (TFs) and global transcriptional consequences of overexpressing 206 TFs. This analysis has discovered specific TFs that mediate conditional co-regulation of genes within 240 modules across 14 distinct environmental contexts. In addition to recapitulating previously characterized regulons, we discovered 454 novel mechanisms for gene regulation during stress, cholesterol utilization and dormancy. Significantly, 183 of these mechanisms act uniquely under conditions experienced during the infection cycle to regulate diverse functions including 23 genes that are essential to host-pathogen interactions. These and other insights underscore the power of a rational, model-driven approach to unearth novel MTB biology that operates under some but not all phases of infection.


Subject(s)
Gene Expression Regulation, Bacterial , Gene Regulatory Networks , Mycobacterium tuberculosis/genetics , Cholesterol/metabolism , Gene Expression Profiling , Genome, Bacterial , Models, Genetic , Transcription Factors/metabolism , Transcription, Genetic
7.
Nucleic Acids Res ; 42(3): 1442-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24185701

ABSTRACT

Systems scale models provide the foundation for an effective iterative cycle between hypothesis generation, experiment and model refinement. Such models also enable predictions facilitating the understanding of biological complexity and the control of biological systems. Here, we demonstrate the reconstruction of a globally predictive gene regulatory model from public data: a model that can drive rational experiment design and reveal new regulatory mechanisms underlying responses to novel environments. Specifically, using ∼ 1500 publically available genome-wide transcriptome data sets from Saccharomyces cerevisiae, we have reconstructed an environment and gene regulatory influence network that accurately predicts regulatory mechanisms and gene expression changes on exposure of cells to completely novel environments. Focusing on transcriptional networks that induce peroxisomes biogenesis, the model-guided experiments allow us to expand a core regulatory network to include novel transcriptional influences and linkage across signaling and transcription. Thus, the approach and model provides a multi-scalar picture of gene dynamics and are powerful resources for exploiting extant data to rationally guide experimentation. The techniques outlined here are generally applicable to any biological system, which is especially important when experimental systems are challenging and samples are difficult and expensive to obtain-a common problem in laboratory animal and human studies.


Subject(s)
Gene Regulatory Networks , Systems Biology/methods , Gene Expression Profiling , Gene Expression Regulation, Fungal , Saccharomyces cerevisiae/genetics
8.
Mol Syst Biol ; 10: 740, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-25028489

ABSTRACT

Microbes can tailor transcriptional responses to diverse environmental challenges despite having streamlined genomes and a limited number of regulators. Here, we present data-driven models that capture the dynamic interplay of the environment and genome-encoded regulatory programs of two types of prokaryotes: Escherichia coli (a bacterium) and Halobacterium salinarum (an archaeon). The models reveal how the genome-wide distributions of cis-acting gene regulatory elements and the conditional influences of transcription factors at each of those elements encode programs for eliciting a wide array of environment-specific responses. We demonstrate how these programs partition transcriptional regulation of genes within regulons and operons to re-organize gene-gene functional associations in each environment. The models capture fitness-relevant co-regulation by different transcriptional control mechanisms acting across the entire genome, to define a generalized, system-level organizing principle for prokaryotic gene regulatory networks that goes well beyond existing paradigms of gene regulation. An online resource (http://egrin2.systemsbiology.net) has been developed to facilitate multiscale exploration of conditional gene regulation in the two prokaryotes.


Subject(s)
Gene Regulatory Networks , Genome, Microbial , Models, Genetic , Algorithms , Escherichia coli/genetics , Gene Expression Regulation , Genetic Fitness , Halobacterium salinarum/genetics , Operon , Regulatory Elements, Transcriptional , Regulon
9.
Nucleic Acids Res ; 41(1): 509-17, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23125364

ABSTRACT

Mycobacterium tuberculosis (MTB) is a highly successful pathogen that infects over a billion people. As with most organisms, MTB adapts to stress by modifying its transcriptional profile. Remodeling of the transcriptome requires both altering the transcription rate and clearing away the existing mRNA through degradation, a process that can be directly regulated in response to stress. To understand better how MTB adapts to the harsh environs of the human host, we performed a global survey of the decay rates of MTB mRNA transcripts. Decay rates were measured for 2139 of the ~4000 MTB genes, which displayed an average half-life of 9.5 min. This is nearly twice the average mRNA half-life of other prokaryotic organisms where these measurements have been made. The transcriptome was further stabilized in response to lowered temperature and hypoxic stress. The generally stable transcriptome described here, and the additional stabilization in response to physiologically relevant stresses, has far-ranging implications for how this pathogen is able to adapt in its human host.


Subject(s)
Mycobacterium tuberculosis/genetics , RNA Stability , RNA, Messenger/metabolism , Cold Temperature , Half-Life , Mycobacterium tuberculosis/metabolism , Stress, Physiological/genetics , Transcriptome
10.
Genome Res ; 21(11): 1892-904, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21750103

ABSTRACT

Assembly of genes into operons is generally viewed as an important process during the continual adaptation of microbes to changing environmental challenges. However, the genome reorganization events that drive this process are also the roots of instability for existing operons. We have determined that there exists a statistically significant trend that correlates the proportion of genes encoded in operons in archaea to their phylogenetic lineage. We have further characterized how microbes deal with operon instability by mapping and comparing transcriptome architectures of four phylogenetically diverse extremophiles that span the range of operon stabilities observed across archaeal lineages: a photoheterotrophic halophile (Halobacterium salinarum NRC-1), a hydrogenotrophic methanogen (Methanococcus maripaludis S2), an acidophilic and aerobic thermophile (Sulfolobus solfataricus P2), and an anaerobic hyperthermophile (Pyrococcus furiosus DSM 3638). We demonstrate how the evolution of transcriptional elements (promoters and terminators) generates new operons, restores the coordinated regulation of translocated, inverted, and newly acquired genes, and introduces completely novel regulation for even some of the most conserved operonic genes such as those encoding subunits of the ribosome. The inverse correlation (r=-0.92) between the proportion of operons with such internally located transcriptional elements and the fraction of conserved operons in each of the four archaea reveals an unprecedented view into varying stages of operon evolution. Importantly, our integrated analysis has revealed that organisms adapted to higher growth temperatures have lower tolerance for genome reorganization events that disrupt operon structures.


Subject(s)
Evolution, Molecular , Genome, Archaeal , Transcriptome , Adenosine Triphosphatases/genetics , Archaea/classification , Archaea/genetics , Gene Expression Profiling , Gene Expression Regulation, Archaeal , Genes, Archaeal , Operon , Phylogeny , Promoter Regions, Genetic , Protein Biosynthesis/genetics , RNA Transport , Transcription, Genetic , Transcriptional Activation
11.
Mol Cancer Ther ; 22(3): 406-418, 2023 03 02.
Article in English | MEDLINE | ID: mdl-36595660

ABSTRACT

In the TRANSCEND NHL 001 study, 53% of patients with relapsed/refractory large B-cell lymphoma (LBCL) treated with lisocabtagene maraleucel (liso-cel) achieved a complete response (CR). To determine characteristics of patients who did and did not achieve a CR, we examined the tumor biology and microenvironment from lymph node tumor biopsies. LBCL biopsies from liso-cel-treated patients were taken pretreatment and ∼11 days posttreatment for RNA sequencing (RNA-seq) and multiplex immunofluorescence (mIF). We analyzed gene expression data from pretreatment biopsies (N = 78) to identify gene sets enriched in patients who achieved a CR to those with progressive disease. Pretreatment biopsies from month-3 CR patients displayed higher expression levels of T-cell and stroma-associated genes, and lower expression of cell-cycle genes. To interpret whether LBCL samples were "follicular lymphoma (FL)-like," we constructed an independent gene expression signature and found that patients with a higher "FL-like" gene expression score had longer progression-free survival (PFS). Cell of origin was not associated with response or PFS, but double-hit gene expression was associated with shorter PFS. The day 11 posttreatment samples (RNA-seq, N = 73; mIF, N = 53) had higher levels of chimeric antigen receptor (CAR) T-cell densities and CAR gene expression, general immune infiltration, and immune activation in patients with CR. Further, the majority of T cells in the day 11 samples were endogenous. Gene expression signatures in liso-cel-treated patients with LBCL can inform the development of combination therapies and next-generation CAR T-cell therapies.


Subject(s)
Lymphoma, Follicular , Lymphoma, Large B-Cell, Diffuse , Receptors, Chimeric Antigen , Humans , Tumor Microenvironment , Biopsy , Genes, Neoplasm , Combined Modality Therapy , Immunotherapy, Adoptive , Antigens, CD19
12.
Cell Rep ; 42(8): 112875, 2023 08 29.
Article in English | MEDLINE | ID: mdl-37542718

ABSTRACT

The success of Mycobacterium tuberculosis (Mtb) is largely attributed to its ability to physiologically adapt and withstand diverse localized stresses within host microenvironments. Here, we present a data-driven model (EGRIN 2.0) that captures the dynamic interplay of environmental cues and genome-encoded regulatory programs in Mtb. Analysis of EGRIN 2.0 shows how modulation of the MtrAB two-component signaling system tunes Mtb growth in response to related host microenvironmental cues. Disruption of MtrAB by tunable CRISPR interference confirms that the signaling system regulates multiple peptidoglycan hydrolases, among other targets, that are important for cell division. Further, MtrA decreases the effectiveness of antibiotics by mechanisms of both intrinsic resistance and drug tolerance. Together, the model-enabled dissection of complex MtrA regulation highlights its importance as a drug target and illustrates how EGRIN 2.0 facilitates discovery and mechanistic characterization of Mtb adaptation to specific host microenvironments within the host.


Subject(s)
Mycobacterium tuberculosis , Transcription Factors , Transcription Factors/genetics , Bacterial Proteins/genetics , Cell Division , Drug Tolerance
13.
Mol Syst Biol ; 7: 554, 2011 Nov 22.
Article in English | MEDLINE | ID: mdl-22108796

ABSTRACT

Numerous lineage-specific expansions of the transcription factor B (TFB) family in archaea suggests an important role for expanded TFBs in encoding environment-specific gene regulatory programs. Given the characteristics of hypersaline lakes, the unusually large numbers of TFBs in halophilic archaea further suggests that they might be especially important in rapid adaptation to the challenges of a dynamically changing environment. Motivated by these observations, we have investigated the implications of TFB expansions by correlating sequence variations, regulation, and physical interactions of all seven TFBs in Halobacterium salinarum NRC-1 to their fitness landscapes, functional hierarchies, and genetic interactions across 2488 experiments covering combinatorial variations in salt, pH, temperature, and Cu stress. This systems analysis has revealed an elegant scheme in which completely novel fitness landscapes are generated by gene conversion events that introduce subtle changes to the regulation or physical interactions of duplicated TFBs. Based on these insights, we have introduced a synthetically redesigned TFB and altered the regulation of existing TFBs to illustrate how archaea can rapidly generate novel phenotypes by simply reprogramming their TFB regulatory network.


Subject(s)
Adaptation, Physiological/genetics , Archaeal Proteins/genetics , Halobacterium salinarum/metabolism , Transcription Factor TFIIB/genetics , Transcription Factor TFIIB/metabolism , Archaeal Proteins/metabolism , Evolution, Molecular , Gene Expression Regulation, Archaeal , Halobacterium salinarum/genetics , Phylogeny , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Salt Tolerance , Stress, Physiological
14.
Bioinformatics ; 26(15): 1879-86, 2010 Aug 01.
Article in English | MEDLINE | ID: mdl-20525821

ABSTRACT

RESULTS: We have developed LeTICE (Learning Transcriptional networks from the Integration of ChIP-chip and Expression data), an algorithm for learning a transcriptional network from ChIP-chip and expression data. The network is specified by a binary matrix of transcription factor (TF)-gene interactions partitioning genes into modules and a background of genes that are not involved in the transcriptional regulation. We define a likelihood of a network, and then search for the network optimizing the likelihood. We applied LeTICE to the location and expression data from yeast cells grown in rich media to learn the transcriptional network specific to the yeast cell cycle. It found 12 condition-specific TFs and 15 modules each of which is highly represented with functions related to particular phases of cell-cycle regulation. AVAILABILITY: Our algorithm is available at http://linus.nci.nih.gov/Data/YounA/LeTICE.zip


Subject(s)
Algorithms , Computational Biology/methods , Gene Regulatory Networks , Cell Cycle/genetics , Cell Cycle/physiology , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation , Oligonucleotide Array Sequence Analysis , Saccharomyces cerevisiae/cytology , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
Mol Syst Biol ; 6: 393, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20664639

ABSTRACT

Complexity of cellular response to oxidative stress (OS) stems from its wide-ranging damage to nucleic acids, proteins, carbohydrates, and lipids. We have constructed a systems model of OS response (OSR) for Halobacterium salinarum NRC-1 in an attempt to understand the architecture of its regulatory network that coordinates this complex response. This has revealed a multi-tiered OS-management program to transcriptionally coordinate three peroxidase/catalase enzymes, two superoxide dismutases, production of rhodopsins, carotenoids and gas vesicles, metal trafficking, and various other aspects of metabolism. Through experimental validation of interactions within the OSR regulatory network, we show that despite their inability to directly sense reactive oxygen species, general transcription factors have an important function in coordinating this response. Remarkably, a significant fraction of this OSR was accurately recapitulated by a model that was earlier constructed from cellular responses to diverse environmental perturbations--this constitutes the general stress response component. Notwithstanding this observation, comparison of the two models has identified the coordination of frontline defense and repair systems by regulatory mechanisms that are triggered uniquely by severe OS and not by other environmental stressors, including sub-inhibitory levels of redox-active metals, extreme changes in oxygen tension, and a sub-lethal dose of gamma rays.


Subject(s)
Archaeal Proteins/metabolism , Halobacterium salinarum/metabolism , Oxidative Stress , Oxygen/metabolism , Reactive Oxygen Species/metabolism , Archaeal Proteins/genetics , Carotenoids/metabolism , Catalase/metabolism , Dose-Response Relationship, Drug , Gene Expression Regulation, Archaeal , Genotype , Halobacterium salinarum/drug effects , Halobacterium salinarum/enzymology , Halobacterium salinarum/genetics , Halobacterium salinarum/growth & development , Hydrogen Peroxide/pharmacology , Models, Biological , Mutation , Oxidants/pharmacology , Oxidation-Reduction , Oxidative Stress/drug effects , Oxidative Stress/genetics , Paraquat/pharmacology , Peroxidases/metabolism , Phenotype , Protein Transport , Rhodopsins, Microbial/metabolism , Superoxide Dismutase/metabolism , Superoxides/metabolism , Time Factors , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 105(24): 8309-14, 2008 Jun 17.
Article in English | MEDLINE | ID: mdl-18550811

ABSTRACT

Cohesin is required to prevent premature dissociation of sister chromatids after DNA replication. Although its role in chromatid cohesion is well established, the functional significance of cohesin's association with interphase chromatin is not clear. Using a quantitative proteomics approach, we show that the STAG1 (Scc3/SA1) subunit of cohesin interacts with the CCTC-binding factor CTCF bound to the c-myc insulator element. Both allele-specific binding of CTCF and Scc3/SA1 at the imprinted IGF2/H19 gene locus and our analyses of human DM1 alleles containing base substitutions at CTCF-binding motifs indicate that cohesin recruitment to chromosomal sites depends on the presence of CTCF. A large-scale genomic survey using ChIP-Chip demonstrates that Scc3/SA1 binding strongly correlates with the CTCF-binding site distribution in chromosomal arms. However, some chromosomal sites interact exclusively with CTCF, whereas others interact with Scc3/SA1 only. Furthermore, immunofluorescence microscopy and ChIP-Chip experiments demonstrate that CTCF associates with both centromeres and chromosomal arms during metaphase. These results link cohesin to gene regulatory functions and suggest an essential role for CTCF during sister chromatid cohesion. These results have implications for the functional role of cohesin subunits in the pathogenesis of Cornelia de Lange syndrome and Roberts syndromes.


Subject(s)
Cell Cycle Proteins/metabolism , Centromere/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , 3T3 Cells , Alleles , Amino Acid Sequence , Animals , CCCTC-Binding Factor , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromatin Immunoprecipitation , Chromosomal Proteins, Non-Histone/genetics , Chromosomes, Human/metabolism , DNA-Binding Proteins/genetics , Genomic Imprinting , Genomics , Humans , Insulator Elements , Insulin-Like Growth Factor II/genetics , Jurkat Cells , Mass Spectrometry , Mice , Molecular Sequence Data , Nuclear Proteins/genetics , Proteomics , Repressor Proteins/genetics , Cohesins
17.
NPJ Precis Oncol ; 5(1): 60, 2021 Jun 28.
Article in English | MEDLINE | ID: mdl-34183722

ABSTRACT

Despite recent advancements in the treatment of multiple myeloma (MM), nearly all patients ultimately relapse and many become refractory to multiple lines of therapies. Therefore, we not only need the ability to predict which patients are at high risk for disease progression but also a means to understand the mechanisms underlying their risk. Here, we report a transcriptional regulatory network (TRN) for MM inferred from cross-sectional multi-omics data from 881 patients that predicts how 124 chromosomal abnormalities and somatic mutations causally perturb 392 transcription regulators of 8549 genes to manifest in distinct clinical phenotypes and outcomes. We identified 141 genetic programs whose activity profiles stratify patients into 25 distinct transcriptional states and proved to be more predictive of outcomes than did mutations. The coherence of these programs and accuracy of our network-based risk prediction was validated in two independent datasets. We observed subtype-specific vulnerabilities to interventions with existing drugs and revealed plausible mechanisms for relapse, including the establishment of an immunosuppressive microenvironment. Investigation of the t(4;14) clinical subtype using the TRN revealed that 16% of these patients exhibit an extreme-risk combination of genetic programs (median progression-free survival of 5 months) that create a distinct phenotype with targetable genes and pathways.

18.
BMC Bioinformatics ; 11: 382, 2010 Jul 19.
Article in English | MEDLINE | ID: mdl-20642854

ABSTRACT

BACKGROUND: High-density tiling arrays and new sequencing technologies are generating rapidly increasing volumes of transcriptome and protein-DNA interaction data. Visualization and exploration of this data is critical to understanding the regulatory logic encoded in the genome by which the cell dynamically affects its physiology and interacts with its environment. RESULTS: The Gaggle Genome Browser is a cross-platform desktop program for interactively visualizing high-throughput data in the context of the genome. Important features include dynamic panning and zooming, keyword search and open interoperability through the Gaggle framework. Users may bookmark locations on the genome with descriptive annotations and share these bookmarks with other users. The program handles large sets of user-generated data using an in-process database and leverages the facilities of SQL and the R environment for importing and manipulating data.A key aspect of the Gaggle Genome Browser is interoperability. By connecting to the Gaggle framework, the genome browser joins a suite of interconnected bioinformatics tools for analysis and visualization with connectivity to major public repositories of sequences, interactions and pathways. To this flexible environment for exploring and combining data, the Gaggle Genome Browser adds the ability to visualize diverse types of data in relation to its coordinates on the genome. CONCLUSIONS: Genomic coordinates function as a common key by which disparate biological data types can be related to one another. In the Gaggle Genome Browser, heterogeneous data are joined by their location on the genome to create information-rich visualizations yielding insight into genome organization, transcription and its regulation and, ultimately, a better understanding of the mechanisms that enable the cell to dynamically respond to its environment.


Subject(s)
Genomics/methods , Systems Biology/methods , Bacillus anthracis/genetics , Gene Expression Profiling , Gene Expression Regulation , Genome, Archaeal , Halobacterium salinarum/genetics , Software
19.
Mol Syst Biol ; 5: 282, 2009.
Article in English | MEDLINE | ID: mdl-19536205

ABSTRACT

During evolution, enzyme-coding genes are acquired and/or replaced through lateral gene transfer and compiled into metabolic pathways. Gene regulatory networks evolve to fine tune biochemical fluxes through such metabolic pathways, enabling organisms to acclimate to nutrient fluctuations in a competitive environment. Here, we demonstrate that a single TrmB family transcription factor in Halobacterium salinarum NRC-1 globally coordinates functionally linked enzymes of diverse phylogeny in response to changes in carbon source availability. Specifically, during nutritional limitation, TrmB binds a cis-regulatory element to activate or repress 113 promoters of genes encoding enzymes in diverse metabolic pathways. By this mechanism, TrmB coordinates the expression of glycolysis, TCA cycle, and amino-acid biosynthesis pathways with the biosynthesis of their cognate cofactors (e.g. purine and thiamine). Notably, the TrmB-regulated metabolic network includes enzyme-coding genes that are uniquely archaeal as well as those that are conserved across all three domains of life. Simultaneous analysis of metabolic and gene regulatory network architectures suggests an ongoing process of co-evolution in which TrmB integrates the expression of metabolic enzyme-coding genes of diverse origins.


Subject(s)
Archaeal Proteins/genetics , Halobacterium salinarum/genetics , Metabolic Networks and Pathways/genetics , Transcription Factors/genetics , Amino Acid Sequence , Archaeal Proteins/metabolism , Binding Sites , Carbohydrate Metabolism , Databases, Genetic , Enhancer Elements, Genetic , Evolution, Molecular , Gene Expression Profiling , Gene Regulatory Networks , Halobacterium salinarum/metabolism , Metabolic Networks and Pathways/physiology , Molecular Sequence Data , Oxidation-Reduction , Phenotype , Promoter Regions, Genetic , Sequence Alignment , Sequence Analysis, Protein , Signal Transduction/genetics , Signal Transduction/physiology , Stress, Physiological , Transcription Factors/metabolism
20.
Mol Syst Biol ; 5: 285, 2009.
Article in English | MEDLINE | ID: mdl-19536208

ABSTRACT

Despite the knowledge of complex prokaryotic-transcription mechanisms, generalized rules, such as the simplified organization of genes into operons with well-defined promoters and terminators, have had a significant role in systems analysis of regulatory logic in both bacteria and archaea. Here, we have investigated the prevalence of alternate regulatory mechanisms through genome-wide characterization of transcript structures of approximately 64% of all genes, including putative non-coding RNAs in Halobacterium salinarum NRC-1. Our integrative analysis of transcriptome dynamics and protein-DNA interaction data sets showed widespread environment-dependent modulation of operon architectures, transcription initiation and termination inside coding sequences, and extensive overlap in 3' ends of transcripts for many convergently transcribed genes. A significant fraction of these alternate transcriptional events correlate to binding locations of 11 transcription factors and regulators (TFs) inside operons and annotated genes-events usually considered spurious or non-functional. Using experimental validation, we illustrate the prevalence of overlapping genomic signals in archaeal transcription, casting doubt on the general perception of rigid boundaries between coding sequences and regulatory elements.


Subject(s)
Genes, Archaeal , Operon , Promoter Regions, Genetic , Transcription Factors/genetics , Computer Simulation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Gene Regulatory Networks , Genome, Bacterial , Halobacterium salinarum/genetics , Halobacterium salinarum/physiology , Models, Genetic , Monte Carlo Method , RNA/genetics , Reproducibility of Results , Transcription Factors/metabolism , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL