Network


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

Hotspot


Dive into the research topics where Adria Carbo is active.

Publication


Featured researches published by Adria Carbo.


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.


Journal of Biological Chemistry | 2011

Abscisic Acid Regulates Inflammation via Ligand-binding Domain-independent Activation of Peroxisome Proliferator-activated Receptor γ

Josep Bassaganya-Riera; Amir J. Guri; Pinyi Lu; Montse Climent; Adria Carbo; Bruno W. S. Sobral; William Horne; Stephanie N. Lewis; David R. Bevan; Raquel Hontecillas

Abscisic acid (ABA) has shown efficacy in the treatment of diabetes and inflammation; however, its molecular targets and the mechanisms of action underlying its immunomodulatory effects remain unclear. This study investigates the role of peroxisome proliferator-activated receptor γ (PPAR γ) and lanthionine synthetase C-like 2 (LANCL2) as molecular targets for ABA. We demonstrate that ABA increases PPAR γ reporter activity in RAW 264.7 macrophages and increases ppar γ expression in vivo, although it does not bind to the ligand-binding domain of PPAR γ. LANCL2 knockdown studies provide evidence that ABA-mediated activation of macrophage PPAR γ is dependent on lancl2 expression. Consistent with the association of LANCL2 with G proteins, we provide evidence that ABA increases cAMP accumulation in immune cells. ABA suppresses LPS-induced prostaglandin E2 and MCP-1 production via a PPAR γ-dependent mechanism possibly involving activation of PPAR γ and suppression of NF-κB and nuclear factor of activated T cells. LPS challenge studies in PPAR γ-expressing and immune cell-specific PPAR γ null mice demonstrate that ABA down-regulates toll-like receptor 4 expression in macrophages and T cells in vivo through a PPAR γ-dependent mechanism. Global transcriptomic profiling and confirmatory quantitative RT-PCR suggest novel candidate targets and demonstrate that ABA treatment mitigates the effect of LPS on the expression of genes involved in inflammation, metabolism, and cell signaling, in part, through PPAR γ. In conclusion, ABA decreases LPS-mediated inflammation and regulates innate immune responses through a bifurcating pathway involving LANCL2 and an alternative, ligand-binding domain-independent mechanism of PPAR γ activation.


British Journal of Nutrition | 2011

Activation of PPARγ and δ by dietary punicic acid ameliorates intestinal inflammation in mice

Josep Bassaganya-Riera; Margaret DiGuardo; Montse Climent; Cristina Vives; Adria Carbo; Zeina Jouni; Alexandra Einerhand; Marianne O'Shea; Raquel Hontecillas

The goal of the present study was to elucidate the mechanisms of immunoregulation by which dietary punicic acid (PUA) prevents or ameliorates experimental inflammatory bowel disease (IBD). The expression of PPARγ and δ, their responsive genes and pro-inflammatory cytokines was assayed in the colonic mucosa. Immune cell-specific PPARγ null, PPARδ knockout and wild-type mice were treated with PUA and challenged with 2·5 % dextran sodium sulphate (DSS). The prophylactic efficacy of PUA was examined in an IL-10(-/-) model of IBD. The effect of PUA on the regulatory T-cell (Treg) compartment was also examined in mice with experimental IBD. PUA ameliorated spontaneous pan-enteritis in IL-10(-/-) mice and DSS colitis, up-regulated Foxp3 expression in Treg and suppressed TNF-α, but the loss of functional PPARγ or δ impaired these anti-inflammatory effects. At the cellular level, the macrophage-specific deletion of PPARγ caused a complete abrogation of the protective effect of PUA, whereas the deletion of PPARδ or intestinal epithelial cell-specific PPARγ decreased its anti-inflammatory efficacy. We provide in vivo molecular evidence demonstrating that PUA ameliorates experimental IBD by regulating macrophage and T-cell function through PPARγ- and δ-dependent mechanisms.


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.


PLOS ONE | 2010

Immunoregulatory Actions of Epithelial Cell PPAR γ at the Colonic Mucosa of Mice with Experimental Inflammatory Bowel Disease

Saroj K. Mohapatra; Amir J. Guri; Montse Climent; Cristina Vives; Adria Carbo; William Horne; Raquel Hontecillas; Josep Bassaganya-Riera

Background Peroxisome proliferator-activated receptors are nuclear receptors highly expressed in intestinal epithelial cells (IEC) and immune cells within the gut mucosa and are implicated in modulating inflammation and immune responses. The objective of this study was to investigate the effect of targeted deletion of PPAR γ in IEC on progression of experimental inflammatory bowel disease (IBD). Methodology/Principal Findings In the first phase, PPAR γ flfl; Villin Cre- (VC-) and PPAR γ flfl; Villin Cre+ (VC+) mice in a mixed FVB/C57BL/6 background were challenged with 2.5% dextran sodium sulfate (DSS) in drinking water for 0, 2, or 7 days. VC+ mice express a transgenic recombinase under the control of the Villin-Cre promoter that causes an IEC-specific deletion of PPAR γ. In the second phase, we generated VC- and VC+ mice in a C57BL/6 background that were challenged with 2.5% DSS. Mice were scored on disease severity both clinically and histopathologically. Flow cytometry was used to phenotypically characterize lymphocyte and macrophage populations in blood, spleen and mesenteric lymph nodes. Global gene expression analysis was profiled using Affymetrix microarrays. The IEC-specific deficiency of PPAR γ in mice with a mixed background worsened colonic inflammatory lesions, but had no effect on disease activity (DAI) or weight loss. In contrast, the IEC-specific PPAR γ null mice in C57BL/6 background exhibited more severe inflammatory lesions, DAI and weight loss in comparison to their littermates expressing PPAR γ in IEC. Global gene expression profiling revealed significantly down-regulated expression of lysosomal pathway genes and flow cytometry results demonstrated suppressed production of IL-10 by CD4+ T cells in mesenteric lymph nodes (MLN) of IEC-specific PPAR γ null mice. Conclusions/Significance Our results demonstrate that adequate expression of PPAR γ in IEC is required for the regulation of mucosal immune responses and prevention of experimental IBD, possibly by modulation of lysosomal and antigen presentation pathways.


PLOS ONE | 2012

Computational Modeling-Based Discovery of Novel Classes of Anti-Inflammatory Drugs That Target Lanthionine Synthetase C-Like Protein 2

Pinyi Lu; Raquel Hontecillas; William Horne; Adria Carbo; Monica Viladomiu; Mireia Pedragosa; David R. Bevan; Stephanie N. Lewis; Josep Bassaganya-Riera

Background Lanthionine synthetase component C-like protein 2 (LANCL2) is a member of the eukaryotic lanthionine synthetase component C-Like protein family involved in signal transduction and insulin sensitization. Recently, LANCL2 is a target for the binding and signaling of abscisic acid (ABA), a plant hormone with anti-diabetic and anti-inflammatory effects. Methodology/Principal Findings The goal of this study was to determine the role of LANCL2 as a potential therapeutic target for developing novel drugs and nutraceuticals against inflammatory diseases. Previously, we performed homology modeling to construct a three-dimensional structure of LANCL2 using the crystal structure of lanthionine synthetase component C-like protein 1 (LANCL1) as a template. Using this model, structure-based virtual screening was performed using compounds from NCI (National Cancer Institute) Diversity Set II, ChemBridge, ZINC natural products, and FDA-approved drugs databases. Several potential ligands were identified using molecular docking. In order to validate the anti-inflammatory efficacy of the top ranked compound (NSC61610) in the NCI Diversity Set II, a series of in vitro and pre-clinical efficacy studies were performed using a mouse model of dextran sodium sulfate (DSS)-induced colitis. Our findings showed that the lead compound, NSC61610, activated peroxisome proliferator-activated receptor gamma in a LANCL2- and adenylate cyclase/cAMP dependent manner in vitro and ameliorated experimental colitis by down-modulating colonic inflammatory gene expression and favoring regulatory T cell responses. Conclusions/Significance LANCL2 is a novel therapeutic target for inflammatory diseases. High-throughput, structure-based virtual screening is an effective computational-based drug design method for discovering anti-inflammatory LANCL2-based drug candidates.


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.


PLOS ONE | 2012

Modeling the Role of Peroxisome Proliferator-Activated Receptor γ and MicroRNA-146 in Mucosal Immune Responses to Clostridium difficile

Monica Viladomiu; Raquel Hontecillas; Mireia Pedragosa; Adria Carbo; Stefan Hoops; Pawel Michalak; Katarzyna Michalak; Richard L. Guerrant; James K. Roche; Cirle A. Warren; Josep Bassaganya-Riera

Clostridium difficile is an anaerobic bacterium that has re-emerged as a facultative pathogen and can cause nosocomial diarrhea, colitis or even death. Peroxisome proliferator-activated receptor (PPAR) γ has been implicated in the prevention of inflammation in autoimmune and infectious diseases; however, its role in the immunoregulatory mechanisms modulating host responses to C. difficile and its toxins remains largely unknown. To characterize the role of PPARγ in C. difficile-associated disease (CDAD), immunity and gut pathology, we used a mouse model of C. difficile infection in wild-type and T cell-specific PPARγ null mice. The loss of PPARγ in T cells increased disease activity and colonic inflammatory lesions following C. difficile infection. Colonic expression of IL-17 was upregulated and IL-10 downregulated in colons of T cell-specific PPARγ null mice. Also, both the loss of PPARγ in T cells and C. difficile infection favored Th17 responses in spleen and colonic lamina propria of mice with CDAD. MicroRNA (miRNA)-sequencing analysis and RT-PCR validation indicated that miR-146b was significantly overexpressed and nuclear receptor co-activator 4 (NCOA4) suppressed in colons of C. difficile-infected mice. We next developed a computational model that predicts the upregulation of miR-146b, downregulation of the PPARγ co-activator NCOA4, and PPARγ, leading to upregulation of IL-17. Oral treatment of C. difficile-infected mice with the PPARγ agonist pioglitazone ameliorated colitis and suppressed pro-inflammatory gene expression. In conclusion, our data indicates that miRNA-146b and PPARγ activation may be implicated in the regulation of Th17 responses and colitis in C. difficile-infected mice.


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.


international parallel and distributed processing symposium | 2012

High-Performance Interaction-Based Simulation of Gut Immunopathologies with ENteric Immunity Simulator (ENISI)

Keith R. Bisset; Md. Maksudul Alam; Josep Bassaganya-Riera; Adria Carbo; Stephen Eubank; Raquel Hontecillas; Stefan Hoops; Yongguo Mei; Katherine Wendelsdorf; Dawen Xie; Jae-Seung Yeom; Madhav V. Marathe

Here we present the ENteric Immunity Simulator (ENISI), a modeling system for the inflammatory and regulatory immune pathways triggered by microbe-immune cell interactions in the gut. With ENISI, immunologists and infectious disease experts can test and generate hypotheses for enteric disease pathology and propose interventions through experimental infection of an in silico gut. ENISI is an agent based simulator, in which individual cells move through the simulated tissues, and engage in context-dependent interactions with the other cells with which they are in contact. The scale of ENISI is unprecedented in this domain, with the ability to simulate 107 cells for 250 simulated days on 576 cores in one and a half hours, with the potential to scale to even larger hardware and problem sizes. In this paper we describe the ENISI simulator for modeling mucosal immune responses to gastrointestinal pathogens. We then demonstrate the utility of ENISI by recreating an experimental infection of a mouse with Helicobacter pylori 26695. The results identify specific processes by which bacterial virulence factors do and do not contribute to pathogenesis associated with H. pylori strain 26695. These modeling results inform general intervention strategies by indicating immunomodulatory mechanisms such as those used in inflammatory bowel disease may be more appropriate therapeutically than directly targeting specific microbial populations through vaccination or by using antimicrobials.

Collaboration


Dive into the Adria Carbo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Casandra Philipson

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

Stefan Hoops

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

Vida Abedi

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pinyi Lu

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

Yongguo Mei

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

Andrew Leber

Virginia Bioinformatics Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge