16 research outputs found

    Systems Biology of Microbiota Metabolites and Adipocyte Transcription Factor Network

    Get PDF
    The overall goal of this research is to understand roles of gut microbiota metabolites and adipocyte transcription factor (TF) network in health and disease by developing systematic analysis methods. As microbiota can perform diverse biotransformation reactions, the spectrum of metabolites present in the gastrointestinal (GI) tract is extremely complex but only a handful of bioactive microbiota metabolites have been identified. We developed a metabolomics workflow that integrates in silico discovery with targeted mass spectrometry. A computational pathway analysis where microbiota metabolisms are modeled as a single metabolic network is utilized to predict a focused set of targets for multiple reaction monitoring (MRM) analysis. We validated our methodology by predicting, quantifying in murine cecum and feces and characterizing tryptophan (TRP)-derived metabolites as ligands for the aryl hydrocarbon receptor. The adipocyte process of lipid droplet accumulation and differentiation is regulated by multiple TFs that function together in a network. Although individual TF activation is previously reported, construction of an integrated network has been limited due to different measurement conditions. We developed an integrated network model of key TFs - PPAR, C/EBP, CREB, NFAT, FoxO1, and SREBP-1c - underlying adipocyte differentiation. A hypothetic model was determined based on literature, and stochastic simulation algorithm (SSA) was applied to simulate TF dynamics. TF activation profiles at different stages of differentiation were measured using 3T3-L1 reporter cell lines where binding of a TF to its DNA binding element drives expression of the Gaussia luciferase gene. Reaction trajectories calculated by SSA showed good agreement with experimental measurement. The TF model was further validated by perturbing dynamics of CREB using forskolin, and comparing the predicted response with experimental data. We studied the molecular recognition mechanism underlying anti-inflammatory function of a bacterial metabolite, indole in DC2.4 cells. The indole treatment attenuated the fraction of cells that were producing the pro-inflammatory cytokine, TNFĪ± and knockdown of nuclear receptor related 1 (Nurr1; NR4A2) resulted in less indole-derived suppression of TNFĪ± production. The first discovery of NR4A2 as a molecular mediator of the endogenous metabolite, indole is expected to provide a new strategy for treatment of inflammatory disorders

    Analysis of Transcription Factor Network Underlying 3T3-L1 Adipocyte Differentiation

    Get PDF
    <div><p>Lipid accumulation in adipocytes reflects a balance between enzymatic pathways leading to the formation and breakdown of esterified lipids, primarily triglycerides. This balance is extremely important, as both high and low lipid levels in adipocytes can have deleterious consequences. The enzymes responsible for lipid synthesis and breakdown (lipogenesis and lipolysis, respectively) are regulated through the coordinated actions of several transcription factors (TFs). In this study, we examined the dynamics of several key transcription factors (TFs) - PPARĪ³, C/EBPĪ², CREB, NFAT, FoxO1, and SREBP-1c - during adipogenic differentiation (week 1) and ensuing lipid accumulation. The activation profiles of these TFs at different times following induction of adipogenic differentiation were quantified using 3T3-L1 reporter cell lines constructed to secrete the <i>Gaussia</i> luciferase enzyme upon binding of a TF to its DNA binding element. The dynamics of the TFs was also modeled using a combination of logical gates and ordinary differential equations, where the logical gates were used to explore different combinations of activating inputs for PPARĪ³, C/EBPĪ², and SREBP-1c. Comparisons of the experimental profiles and model simulations suggest that SREBP-1c could be independently activated by either insulin or PPARĪ³, whereas PPARĪ³ activation required both C/EBPĪ² as well as a putative ligand. Parameter estimation and sensitivity analysis indicate that feedback activation of SREBP-1c by PPARĪ³ is negligible in comparison to activation of SREBP-1c by insulin. On the other hand, the production of an activating ligand could quantitatively contribute to a sustained elevation in PPARĪ³ activity.</p></div

    Simulated activity profiles for (A) CREB, (B) C/EBP, (C) PPARĪ³, and (D) SREBP-1c generated using the top five Hill equation models.

    No full text
    <p>The model numbers are shown in the figure legend, listed in order of increasing sum of squared residuals (SSR). The measured data (normalized mean Gluc activities, RFL/h/RLU) are shown as red dots. The specific combination of logic gates for these models can be determined from <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0100177#pone.0100177.s007" target="_blank">Tables S3</a> and <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0100177#pone.0100177.s008" target="_blank">S4</a>.</p

    Simulated activity profiles for (A) CREB, (B) C/EBP, and (C) PPARĪ³ generated using the best fitting mass action model with added forskolin input.

    No full text
    <p>Forskolin treatment was modeled as a step increase in IBMX during the first induction period (days 0 to 2). All other model parameters (<i>k<sub>1</sub></i>ā€“<i>k<sub>13</sub></i>) were kept at the same values that were estimated from the training data without forskolin. Dashed lines show 95% confidence intervals. The measured data (normalized mean Gluc activities, RFL/h/RLU) are shown as red dots.</p

    Sum of squared residuals (SSR) and adjusted R<sup>2</sup> values for the best ten mass action and Hill equation models representing different logical gate combinations.

    No full text
    <p>Sum of squared residuals (SSR) and adjusted R<sup>2</sup> values for the best ten mass action and Hill equation models representing different logical gate combinations.</p

    Schematic of TF network model.

    No full text
    <p>Arrows indicate direction of interaction. Model parameters labeling the dotted arrows (k<sub>2</sub>, k<sub>5</sub>, k<sub>8</sub>, and k<sub>10</sub>) represent first-order decay rate constants for the TFs. The rate constants shown in the schematic refer to the mass action models. See text for abbreviations.</p

    Simulated activity profiles for (A) CREB, (B) C/EBP, (C) PPARĪ³, and (D) SREBP-1c generated using the top five mass action models.

    No full text
    <p>The model numbers are shown in the figure legend, listed in order of increasing sum of squared residuals (SSR). The measured data (normalized mean Gluc activities, RFL/h/RLU) are shown as red dots. The specific combination of logic gates for these models can be determined from <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0100177#pone.0100177.s007" target="_blank">Tables S3</a> and <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0100177#pone.0100177.s008" target="_blank">S4</a>.</p

    Validation of TF reporter constructs.

    No full text
    <p>(A) Foxo1, CREB, SREBP-1c, and NFAT3 reporter plasmids were validated by co-transfecting plasmids for constitutive expression of each TFs (pCMV5-FLAG-FoxO1, pCMV-Sport6-CREB, pSV Sport SREBP-1c, pEGFP-C1 NFAT3) and corresponding reporter plasmids into 293T/17 cells, and monitoring the TF-mediated Gaussia luciferase (Gluc) activity. A yellow fluorescence protein expressing plasmid (pEYFP-N1) plasmid was co-transfected with each reporter plasmid as a control. (B) The PPARĪ³ reporter construct was verified by transfecting a plasmid containing the PPARĪ³ gene was into 3T3-L1 PPARĪ³ reporter cells, and activating PPARĪ³ with 25 ĀµM of rosiglitazone (RGZ). (C) The C/EBPĪ² reporter construct was validated by activating C/EBPĪ² in 3T3-L1 preadipocyte C/EBPĪ² reporter cells with the cytokine oncostatin M (OSM). Data represent mean Ā± SD. *: p<0.05.</p
    corecore