Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Yongguo Mei is active.

Publication


Featured researches published by Yongguo Mei.


PLOS Computational Biology | 2013

Systems modeling of molecular mechanisms controlling cytokine-driven CD4+ T cell differentiation and phenotype plasticity.

Adria Carbo; Raquel Hontecillas; Barbara Kronsteiner; Monica Viladomiu; Mireia Pedragosa; Pinyi Lu; Casandra Philipson; Stefan Hoops; Madhav V. Marathe; Stephen Eubank; Keith R. Bisset; Katherine Wendelsdorf; Abdul Salam Jarrah; Yongguo Mei; Josep Bassaganya-Riera

Differentiation of CD4+ T cells into effector or regulatory phenotypes is tightly controlled by the cytokine milieu, complex intracellular signaling networks and numerous transcriptional regulators. We combined experimental approaches and computational modeling to investigate the mechanisms controlling differentiation and plasticity of CD4+ T cells in the gut of mice. Our computational model encompasses the major intracellular pathways involved in CD4+ T cell differentiation into T helper 1 (Th1), Th2, Th17 and induced regulatory T cells (iTreg). Our modeling efforts predicted a critical role for peroxisome proliferator-activated receptor gamma (PPARγ) in modulating plasticity between Th17 and iTreg cells. PPARγ regulates differentiation, activation and cytokine production, thereby controlling the induction of effector and regulatory responses, and is a promising therapeutic target for dysregulated immune responses and inflammation. Our modeling efforts predict that following PPARγ activation, Th17 cells undergo phenotype switch and become iTreg cells. This prediction was validated by results of adoptive transfer studies showing an increase of colonic iTreg and a decrease of Th17 cells in the gut mucosa of mice with colitis following pharmacological activation of PPARγ. Deletion of PPARγ in CD4+ T cells impaired mucosal iTreg and enhanced colitogenic Th17 responses in mice with CD4+ T cell-induced colitis. Thus, for the first time we provide novel molecular evidence in vivo demonstrating that PPARγ in addition to regulating CD4+ T cell differentiation also plays a major role controlling Th17 and iTreg plasticity in the gut mucosa.


PLOS ONE | 2013

Predictive computational modeling of the mucosal immune responses during Helicobacter pylori infection.

Adria Carbo; Josep Bassaganya-Riera; Mireia Pedragosa; Monica Viladomiu; Madhav V. Marathe; Stephen Eubank; Katherine Wendelsdorf; Keith R. Bisset; Stefan Hoops; Xinwei Deng; Maksudul Alam; Barbara Kronsteiner; Yongguo Mei; Raquel Hontecillas

T helper (Th) cells play a major role in the immune response and pathology at the gastric mucosa during Helicobacter pylori infection. There is a limited mechanistic understanding regarding the contributions of CD4+ T cell subsets to gastritis development during H. pylori colonization. We used two computational approaches: ordinary differential equation (ODE)-based and agent-based modeling (ABM) to study the mechanisms underlying cellular immune responses to H. pylori and how CD4+ T cell subsets influenced initiation, progression and outcome of disease. To calibrate the model, in vivo experimentation was performed by infecting C57BL/6 mice intragastrically with H. pylori and assaying immune cell subsets in the stomach and gastric lymph nodes (GLN) on days 0, 7, 14, 30 and 60 post-infection. Our computational model reproduced the dynamics of effector and regulatory pathways in the gastric lamina propria (LP) in silico. Simulation results show the induction of a Th17 response and a dominant Th1 response, together with a regulatory response characterized by high levels of mucosal Treg) cells. We also investigated the potential role of peroxisome proliferator-activated receptor γ (PPARγ) activation on the modulation of host responses to H. pylori by using loss-of-function approaches. Specifically, in silico results showed a predominance of Th1 and Th17 cells in the stomach of the cell-specific PPARγ knockout system when compared to the wild-type simulation. Spatio-temporal, object-oriented ABM approaches suggested similar dynamics in induction of host responses showing analogous T cell distributions to ODE modeling and facilitated tracking lesion formation. In addition, sensitivity analysis predicted a crucial contribution of Th1 and Th17 effector responses as mediators of histopathological changes in the gastric mucosa during chronic stages of infection, which were experimentally validated in mice. These integrated immunoinformatics approaches characterized the induction of mucosal effector and regulatory pathways controlled by PPARγ during H. pylori infection affecting disease outcomes.


Frontiers in Cell and Developmental Biology | 2014

Computational modeling of heterogeneity and function of CD4+ T cells

Adria Carbo; Raquel Hontecillas; Tricity Andrew; Kristin Eden; Yongguo Mei; Stefan Hoops; Josep Bassaganya-Riera

The immune system is composed of many different cell types and hundreds of intersecting molecular pathways and signals. This large biological complexity requires coordination between distinct pro-inflammatory and regulatory cell subsets to respond to infection while maintaining tissue homeostasis. CD4+ T cells play a central role in orchestrating immune responses and in maintaining a balance between pro- and anti- inflammatory responses. This tight balance between regulatory and effector reactions depends on the ability of CD4+ T cells to modulate distinct pathways within large molecular networks, since dysregulated CD4+ T cell responses may result in chronic inflammatory and autoimmune diseases. The CD4+ T cell differentiation process comprises an intricate interplay between cytokines, their receptors, adaptor molecules, signaling cascades and transcription factors that help delineate cell fate and function. Computational modeling can help to describe, simulate, analyze, and predict some of the behaviors in this complicated differentiation network. This review provides a comprehensive overview of existing computational immunology methods as well as novel strategies used to model immune responses with a particular focus on CD4+ T cell differentiation.


BMC Bioinformatics | 2015

Multiscale modeling of mucosal immune responses.

Yongguo Mei; Vida Abedi; Adria Carbo; Xiaoying Zhang; Pinyi Lu; Casandra Philipson; Raquel Hontecillas; Stefan Hoops; Nathan Liles; Josep Bassaganya-Riera

Computational modeling techniques are playing increasingly important roles in advancing a systems-level mechanistic understanding of biological processes. Computer simulations guide and underpin experimental and clinical efforts. This study presents ENteric Immune Simulator (ENISI), a multiscale modeling tool for modeling the mucosal immune responses. ENISIs modeling environment can simulate in silico experiments from molecular signaling pathways to tissue level events such as tissue lesion formation. ENISIs architecture integrates multiple modeling technologies including ABM (agent-based modeling), ODE (ordinary differential equations), SDE (stochastic modeling equations), and PDE (partial differential equations). This paper focuses on the implementation and developmental challenges of ENISI. A multiscale model of mucosal immune responses during colonic inflammation, including CD4+ T cell differentiation and tissue level cell-cell interactions was developed to illustrate the capabilities, power and scope of ENISI MSM.BackgroundComputational techniques are becoming increasingly powerful and modeling tools for biological systems are of greater needs. Biological systems are inherently multiscale, from molecules to tissues and from nano-seconds to a lifespan of several years or decades. ENISI MSM integrates multiple modeling technologies to understand immunological processes from signaling pathways within cells to lesion formation at the tissue level. This paper examines and summarizes the technical details of ENISI, from its initial version to its latest cutting-edge implementation.ImplementationObject-oriented programming approach is adopted to develop a suite of tools based on ENISI. Multiple modeling technologies are integrated to visualize tissues, cells as well as proteins; furthermore, performance matching between the scales is addressed.ConclusionWe used ENISI MSM for developing predictive multiscale models of the mucosal immune system during gut inflammation. Our modeling predictions dissect the mechanisms by which effector CD4+ T cell responses contribute to tissue damage in the gut mucosa following immune dysregulation.


bioinformatics and biomedicine | 2012

ENISI Visual, an agent-based simulator for modeling gut immunity

Yongguo Mei; Raquel Hontecillas; Xiaoying Zhang; Keith R. Bisset; Stephen Eubank; Stefan Hoops; Madhav V. Marathe; Josep Bassaganya-Riera

This paper presents ENISI Visual, an agent-based simulator for modeling gut immunity to enteric pathogens. Gastrointestinal systems are important for in-taking food and other nutritions and gut immunity is an important part of human immune system. ENISI Visual provides quality visualizations and users can control initial cell concentrations and the simulation speed, take snapshots, and record videos. The cells are represented with different icons and the icons change colors as their states change. Users can observe real-time immune responses, including cell recruitment, cytokine and chemokine secretion and dissipation, random or chemotactic movement, cell-cell interactions, and state changes. The case study clearly shows that users can use ENISI Visual to develop models and run novel and insightful in silico experiments.


bioinformatics and biomedicine | 2014

ENISI MSM: A novel multi-scale modeling platform for computational immunology

Yongguo Mei; Adria Carbo; Raquel Hontecillas; Stefan Hoops; Nathan Liles; Pinyi Lu; Casandra Philipson; Josep Bassaganya-Riera

Biological systems span several orders of magnitude in space and time from intracellular pathways to tissue-level processes. Many studies focus on molecular level events while other studies focus on cellular level and tissue level interactions. The immune system is highly complex and dynamic, encompassing hierarchical interactions with dimensions ranging from nanometers to meters and time scales from nanoseconds to years. To comprehensively model immunological processes computationally, multi-scale models are needed. However, the lack of multi-scale modeling tools can be a deterrent to advance the understanding of the immune system across scales. In this paper, we developed an object-oriented multi-scale modeling (MSM) platform, ENISI MSM, that integrates agent-based modeling (ABM), ordinary-differential equations (ODE), and partial differential equations (PDE) models. To our best knowledge, this is the first such multi-scale modeling platform that is capable of integrating ODE, PDE, and ABM models together. The tool is developed in Java and is object-oriented. For example, the agents are objects and the ODE and PDE solvers are also objects. ENISI MSM also provides user-friendly interfaces and visualizations. We developed a multi-scale CD4+ T cell differentiation model in the context of gut inflammatory and showed the effectiveness of ENISI MSM.


Biodata Mining | 2015

Supervised learning methods in modeling of CD4+ T cell heterogeneity.

Pinyi Lu; Vida Abedi; Yongguo Mei; Raquel Hontecillas; Stefan Hoops; Adria Carbo; Josep Bassaganya-Riera

BackgroundModeling of the immune system – a highly non-linear and complex system – requires practical and efficient data analytic approaches. The immune system is composed of heterogeneous cell populations and hundreds of cell types, such as neutrophils, eosinophils, macrophages, dendritic cells, T cells, and B cells. Each cell type is highly diverse and can be further differentiated into subsets with unique and overlapping functions. For example, CD4+ T cells can be differentiated into Th1, Th2, Th17, Th9, Th22, Treg, Tfh, as well as Tr1. Each subset plays different roles in the immune system. To study molecular mechanisms of cell differentiation, computational systems biology approaches can be used to represent these processes; however, the latter often requires building complex intracellular signaling models with a large number of equations to accurately represent intracellular pathways and biochemical reactions. Furthermore, studying the immune system entails integration of complex processes which occur at different time and space scales.MethodsThis study presents and compares four supervised learning methods for modeling CD4+ T cell differentiation: Artificial Neural Networks (ANN), Random Forest (RF), Support Vector Machines (SVM), and Linear Regression (LR). Application of supervised learning methods could reduce the complexity of Ordinary Differential Equations (ODEs)-based intracellular models by only focusing on the input and output cytokine concentrations. In addition, this modeling framework can be efficiently integrated into multiscale models.ResultsOur results demonstrate that ANN and RF outperform the other two methods. Furthermore, ANN and RF have comparable performance when applied to in silico data with and without added noise. The trained models were also able to reproduce dynamic behavior when applied to experimental data; in four out of five cases, model predictions based on ANN and RF correctly predicted the outcome of the system. Finally, the running time of different methods was compared, which confirms that ANN is considerably faster than RF.ConclusionsUsing machine learning as opposed to ODE-based method reduces the computational complexity of the system and allows one to gain a deeper understanding of the complex interplay between the different related entities.


bioinformatics and biomedicine | 2013

Neural network models for classifying immune cell subsets

Yongguo Mei; Raquel Hontecillas; Xiaoying Zhang; Adria Carbo; Josep Bassaganya-Riera

The immune system is composed of heterogeneous cell populations and it includes several hundreds of distinct cells types such as neutrohils, eosinophils, basophils, macrophages, dendritic cells, CD4+ and CD8+ T cells, γδ T cells, mast cells, and B cells and each main cell type can be further differentiated into subsets with unique and overlapping functions. For example CD4+ T cells can be differentiated into T helper (Th)1, Th2, Thl7, and regulatory T cell (Treg) subsets. To study molecular mechanisms of cell differentiation, Systems Biology Markup Language (SBML) based Ordinary Differential Equation (ODE) models can be used for representing such processes. These intracellular signaling models often require many equations to accurately represent intracellular pathways and biochemical reactions. Another challenge in studying the immune system and immune responses is the need for integration of complex processes that occur at different time and space scales (i.e., populations, whole organism, tissue level, cellular and molecular) through multi-scale modeling. This study presents two novel neural network models for modeling CD4+ T cell differentiation and immune cell subset classification. The first model reduces the complex ODE intracellular model by focusing on the input and output cytokines and the second model establishes an automated subset classification based on molecular patterns expressed in immune cells. After training, the first model achieves small prediction errors of cytokine concentrations and the second model achieves 98% prediction rate for subset classification. Neural network algorithm and models have been widely used for many data mining tasks such as classification and pattern recognition. However, to the best of our knowledge this study is the first one applying the neural network algorithm for immune cell differentiation and subset classification. In addition, these novel neural network models can be computationally efficiently integrated into multi-scale models with limited computational costs.


PLOS ONE | 2015

Sensitivity Analysis of an ENteric Immunity SImulator (ENISI)-Based Model of Immune Responses to Helicobacter pylori Infection

Maksudul Alam; Xinwei Deng; Casandra Philipson; Josep Bassaganya-Riera; Keith R. Bisset; Adria Carbo; Stephen Eubank; Raquel Hontecillas; Stefan Hoops; Yongguo Mei; Vida Abedi; Madhav V. Marathe

Agent-based models (ABM) are widely used to study immune systems, providing a procedural and interactive view of the underlying system. The interaction of components and the behavior of individual objects is described procedurally as a function of the internal states and the local interactions, which are often stochastic in nature. Such models typically have complex structures and consist of a large number of modeling parameters. Determining the key modeling parameters which govern the outcomes of the system is very challenging. Sensitivity analysis plays a vital role in quantifying the impact of modeling parameters in massively interacting systems, including large complex ABM. The high computational cost of executing simulations impedes running experiments with exhaustive parameter settings. Existing techniques of analyzing such a complex system typically focus on local sensitivity analysis, i.e. one parameter at a time, or a close “neighborhood” of particular parameter settings. However, such methods are not adequate to measure the uncertainty and sensitivity of parameters accurately because they overlook the global impacts of parameters on the system. In this article, we develop novel experimental design and analysis techniques to perform both global and local sensitivity analysis of large-scale ABMs. The proposed method can efficiently identify the most significant parameters and quantify their contributions to outcomes of the system. We demonstrate the proposed methodology for ENteric Immune SImulator (ENISI), a large-scale ABM environment, using a computational model of immune responses to Helicobacter pylori colonization of the gastric mucosa.


IEEE/ACM Transactions on Computational Biology and Bioinformatics | 2015

ENISI SDE: a new web-based tool for modeling stochastic processes

Yongguo Mei; Adria Carbo; Stefan Hoops; Raquel Hontecillas; Josep Bassaganya-Riera

Modeling and simulations approaches have been widely used in computational biology, mathematics, bioinformatics and engineering to represent complex existing knowledge and to effectively generate novel hypotheses. While deterministic modeling strategies are widely used in computational biology, stochastic modeling techniques are not as popular due to a lack of user-friendly tools. This paper presents ENISI SDE, a novel web-based modeling tool with stochastic differential equations. ENISI SDE provides user-friendly web user interfaces to facilitate adoption by immunologists and computational biologists. This work provides three major contributions: (1) discussion of SDE as a generic approach for stochastic modeling in computational biology; (2) development of ENISI SDE, a web-based user-friendly SDE modeling tool that highly resembles regular ODE-based modeling; (3) applying ENISI SDE modeling tool through a use case for studying stochastic sources of cell heterogeneity in the context of CD4+ T cell differentiation. The CD4+ T cell differential ODE model has been published [8] and can be downloaded from biomodels.net. The case study reproduces a biological phenomenon that is not captured by the previously published ODE model and shows the effectiveness of SDE as a stochastic modeling approach in biology in general and immunology in particular and the power of ENISI SDE.

Collaboration


Dive into the Yongguo Mei's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Stefan Hoops

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Casandra Philipson

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vida Abedi

Virginia Bioinformatics Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge