Network


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

Hotspot


Dive into the research topics where Raquel Hontecillas is active.

Publication


Featured researches published by Raquel Hontecillas.


Journal of Immunology | 2007

Peroxisome Proliferator-Activated Receptor γ Is Required for Regulatory CD4+ T Cell-Mediated Protection against Colitis

Raquel Hontecillas; Josep Bassaganya-Riera

Peroxisome proliferator-activated receptor (PPAR) γ activation has been implicated in the prevention of immunoinflammatory disorders; however, the mechanisms of regulation of effector and regulatory CD4+ T cell functions by endogenously activated PPAR-γ remain unclear. We have used PPAR-γ-deficient CD4+ T cells obtained from tissue-specific PPAR-γ null mice (i.e., PPAR-γ fl/fl; MMTV-Cre+) to investigate the role of endogenous PPAR-γ on regulatory T cell (Treg) and effector CD4+ T cell function. Overall, we show that the loss of PPAR-γ results in enhanced Ag-specific proliferation and overproduction of IFN-γ in response to IL-12. These findings correlate in vivo with enhanced susceptibility of tissue-specific PPAR-γ null mice to trinitrobenzene sulfonic acid-induced colitis. Furthermore, the transfer of purified PPAR-γ null CD4+ T cells into SCID recipients results in enteric disease. To test the assertion that the deficiency of PPAR-γ in Treg impairs their ability to prevent effector T cell-induced colitis, we performed cotransfer studies. These studies demonstrate that PPAR-γ-expressing, but not PPAR-γ null Treg, prevent colitis induced by transfer of naive CD4+ T cells into SCID recipients. In line with these findings, the production of IFN-γ by spleen and mesenteric lymph node-derived CD4+ T cells was down-regulated following transfer of PPAR-γ-expressing, but not PPAR-γ null, Treg. In conclusion, our data suggest that endogenous PPAR-γ activation represents a Treg intrinsic mechanism of down-regulation of effector CD4+ T cell function and prevention of colitis.


Journal of The American College of Nutrition | 2009

Activation of PPAR γ and α by punicic acid ameliorates glucose tolerance and suppresses obesity-related inflammation.

Raquel Hontecillas; Marianne O'Shea; Alexandra Einerhand; Margaret DiGuardo; Josep Bassaganya-Riera

Objective: Peroxisome proliferator-activated receptor γ (PPAR γ) is the molecular target for thiazolidinediones (TZDs), a class of synthetic antidiabetic agents. However, the naturally occurring agonists of PPARs remain largely unknown. Punicic acid (PUA) is a conjugated linolenic acid isomer found in pomegrante. The objective of this study was to test the hypothesis that PUA activates PPAR γ and thereby ameliorates glucose homeostasis and obesity-related inflammation. Methods: The ability of PUA to modulate PPAR reporter activity was determined in 3T3-L1 pre-adipocytes. A cell-free assay was used to measure PUAs binding to the ligand-binding domain (LBD) of human PPAR γ. The preventive actions of PUA were investigated using genetically obese db/db mice and a model of diet-induced obesity in PPAR γ-expressing and tissue-specific PPAR γ null mice. Expression of PPAR α, γ, PPAR-responsive genes and TNF-α was measured in tissues controlling glucose homeostasis. Results: PUA caused a dose-dependent increase PPAR α and γ reporter activity in 3T3-L1 cells and bound although weakly to the LBD of human PPAR γ. Dietary PUA decreased fasting plasma glucose concentrations, improved the glucose-normalizing ability, suppressed NF-κB activation, TNF-α expression and upregulated PPAR α- and γ-responsive genes in skeletal muscle and adipose tissue. Loss of PPAR γ impaired the ability of dietary PUA to improve glucose homeostasis and suppress inflammation. Conclusions: Our studies demonstrate that PUA binds and robustly activates PPAR γ, increases PPAR γ-responsive gene expression and the loss of PPAR γ in immune cells impairs its ability to ameliorate diabetes and inflammation.


Journal of Nutritional Biochemistry | 2013

Nutritional protective mechanisms against gut inflammation

Monica Viladomiu; Raquel Hontecillas; Lijuan Yuan; Pinyi Lu; Josep Bassaganya-Riera

Inflammatory bowel disease (IBD) is a debilitating and widespread immune-mediated illness characterized by excessive inflammatory and effector mucosal responses leading to tissue destruction at the gastrointestinal tract. Interactions among the immune system, the commensal microbiota and the host genotype are thought to underlie the pathogenesis of IBD. However, the precise etiology of IBD remains unknown. Diet-induced changes in the composition of the gut microbiome can modulate the induction of regulatory versus effector immune responses at the gut mucosa and improve health outcomes. Therefore, manipulation of gut microbiota composition and the local production of microbial-derived metabolites by using prebiotics, probiotics and dietary fibers is being explored as a promising avenue of prophylactic and therapeutic intervention against gut inflammation. Prebiotics and fiber carbohydrates are fermented by resident microflora into short chain fatty acids (SCFAs) in the colon. SCFAs then activate peroxisome proliferator-activated receptor (PPAR)γ, a nuclear transcription factor with widely demonstrated anti-inflammatory efficacy in experimental IBD. The activation of PPARγ by naturally ocurring compounds such as conjugated linoleic acid, pomegranate seed oil-derived punicic acid, eleostearic acid and abscisic acid has been explored as nutritional interventions that suppress colitis by directly modulating the host immune response. The aim of this review is to summarize the status of innovative nutritional interventions against gastrointestinal inflammation, their proposed mechanisms of action, preclinical and clinical efficacy as well as bioinformatics and computational modeling approaches that accelerate discovery in nutritional and mucosal immunology research.


Journal of Nutrition | 2010

Conjugated Linoleic Acid Ameliorates Inflammation-Induced Colorectal Cancer in Mice through Activation of PPARγ

Nicholas P. Evans; Sarah Misyak; Eva M. Schmelz; Amir J. Guri; Raquel Hontecillas; Josep Bassaganya-Riera

Conjugated linoleic acid (CLA) exerts a protective effect on experimental inflammatory bowel disease and shows promise as a chemopreventive agent against colorectal cancer (CRC) in mice, although the mechanisms by which it exerts its beneficial effects against malignancies in the gut are not completely understood. Mice lacking PPARgamma in immune and epithelial cells and PPARgamma-expressing littermates were fed either control or CLA-supplemented (1 g CLA/100 g) diets to determine the role of PPARgamma in inflammation-induced CRC. To induce tumor formation and colitis, mice were treated with azoxymethane and then challenged with 2% dextran sodium sulfate, respectively. Dietary CLA ameliorated disease activity, decreased colitis, and prevented adenocarcinoma formation in the PPARgamma-expressing floxed mice but not in the tissue-specific PPARgamma-null mice. Dietary CLA supplementation significantly decreased the percentages of macrophages in the mesenteric lymph nodes (MLN) regardless of the genotype and increased regulatory T cell numbers in MLN of PPARgamma-expressing, but not in the tissue-specific, PPARgamma-null mice. Colonic tumor necrosis factor-alpha mRNA expression was significantly suppressed in CLA-fed, PPARgamma-expressing mice. This study suggests CLA ameliorates colitis and prevents tumor formation in part through a PPARgamma-dependent mechanism.


PLOS ONE | 2012

Probiotic Bacteria Produce Conjugated Linoleic Acid Locally in the Gut That Targets Macrophage PPAR γ to Suppress Colitis

Josep Bassaganya-Riera; Monica Viladomiu; Mireia Pedragosa; Claudio De Simone; Adria Carbo; Rustem Shaykhutdinov; Christian Jobin; Janelle C. Arthur; Benjamin A. Corl; Hans J. Vogel; Martin Storr; Raquel Hontecillas

Background Inflammatory bowel disease (IBD) therapies are modestly successful and associated with significant side effects. Thus, the investigation of novel approaches to prevent colitis is important. Probiotic bacteria can produce immunoregulatory metabolites in vitro such as conjugated linoleic acid (CLA), a polyunsaturated fatty acid with potent anti-inflammatory effects. This study aimed to investigate the cellular and molecular mechanisms underlying the anti-inflammatory efficacy of probiotic bacteria using a mouse model of colitis. Methodology/Principal Findings The immune modulatory mechanisms of VSL#3 probiotic bacteria and CLA were investigated in a mouse model of DSS colitis. Colonic specimens were collected for histopathology, gene expression and flow cytometry analyses. Immune cell subsets in the mesenteric lymph nodes (MLN), spleen, blood and colonic lamina propria cells were phenotypically and functionally characterized. Fecal samples and colonic contents were collected to determine the effect of VSL#3 and CLA on gut microbial diversity and CLA production. CLA and VSL#3 treatment ameliorated colitis and decreased colonic bacterial diversity, a finding that correlated with decreased gut pathology. Colonic CLA concentrations were increased in response to probiotic bacterial treatment, but without systemic distribution in blood. VSL#3 and CLA decreased macrophage accumulation in the MLN of mice with DSS colitis. The loss of PPAR γ in myeloid cells abrogated the protective effect of probiotic bacteria and CLA in mice with DSS colitis. Conclusions/Significance Probiotic bacteria modulate gut microbial diversity and favor local production of CLA in the colon that targets myeloid cell PPAR γ to suppress colitis.


Current Opinion in Clinical Nutrition and Metabolic Care | 2010

Dietary conjugated linoleic acid and n-3 polyunsaturated fatty acids in inflammatory bowel disease.

Josep Bassaganya-Riera; Raquel Hontecillas

Purpose of reviewInflammatory bowel disease (IBD) is a debilitating and widespread immune-mediated illness of unknown etiology. Current treatments are modestly successful and with significant side-effects. The purpose of this review is to summarize the current understanding of mechanisms of action underlying the anti-inflammatory actions of conjugated linoleic acid (CLA) and n-3 polyunsaturated fatty acids (PUFAs) in IBD. Recent findingsNutrition-based interventions that target peroxisome proliferator-activated receptors (PPARs) such as dietary CLA and n-3 PUFA have demonstrated anti-inflammatory efficacy in animal models of IBD. Clinical data on n-3 PUFA in IBD remains generally unimpressive, although results of a recent human study demonstrate that IBD remission can be maintained by maintaining the n-3: n-6 ratio more than 0.65 via n-3 PUFA intervention. In mice, CLA prevented inflammation-driven colorectal cancer by activating PPAR γ and modulating regulatory T cells and macrophages. CLA is the subject of an ongoing clinical study in Crohns disease patients. SummaryCompelling evidence demonstrates that n-3 PUFA and CLA prevent or ameliorate IBD in animal models. However, this basic knowledge has not been translated into novel nutrition-based clinical interventions. For both compounds there is an urgent need for placebo-controlled, large-scale, multicenter clinical trials.


Food and Chemical Toxicology | 2011

Pomegranate seed oil, a rich source of punicic acid, prevents diet-induced obesity and insulin resistance in mice

Irene O.C.M. Vroegrijk; Janna A. van Diepen; Sjoerd A. A. van den Berg; Irene Westbroek; Hiskias Keizer; Luisa Gambelli; Raquel Hontecillas; Josep Bassaganya-Riera; Gerben Zondag; Johannes A. Romijn; Louis M. Havekes; Peter J. Voshol

BACKGROUND Pomegranate seed oil has been shown to protect against diet induced obesity and insulin resistance. OBJECTIVE To characterize the metabolic effects of punicic acid on high fat diet induced obesity and insulin resistance. DESIGN High-fat diet or high-fat diet with 1% Pomegranate seed oil (PUA) was fed for 12 weeks to induce obesity and insulin resistance. We assessed body weight and composition (pSABRE DEXA-scan), energy expenditure (Columbus Instruments) and insulin sensitivity at the end of the 12 weeks. RESULTS PSO intake resulted in a lower body weight, 30.5±2.9 vs 33.8±3.2 g PSO vs HFD respectively, p=0.02, without affecting food intake or energy expenditure. The lower body weight was fully explained by a decreased body fat mass, 3.3±2.3 vs 6.7±2.7 g for PSO and HFD fed mice, respectively, p=0.02. Insulin clamps showed that PSO did not affect liver insulin sensitivity but clearly improved peripheral insulin sensitivity, 164±52% vs 92±24% for PSO and HFD fed mice respectively, p=0.01. CONCLUSIONS We conclude that dietary PSO ameliorates high-fat diet induced obesity and insulin resistance in mice, independent of changes in food intake or energy expenditure.


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 | 2012

Immunoregulatory Mechanisms Underlying Prevention of Colitis-Associated Colorectal Cancer by Probiotic Bacteria

Josep Bassaganya-Riera; Monica Viladomiu; Mireia Pedragosa; Claudio De Simone; Raquel Hontecillas

Background Inflammatory bowel disease (IBD) increases the risk of colorectal cancer. Probiotic bacteria produce immunoregulatory metabolites in vitro such as conjugated linoleic acid (CLA), a polyunsaturated fatty acid with potent anti-carcinogenic effects. This study aimed to investigate the cellular and molecular mechanisms underlying the efficacy of probiotic bacteria in mouse models of cancer. Methodology/Principal Findings The immune modulatory mechanisms of VSL#3 probiotic bacteria and CLA were investigated in mouse models of inflammation-driven colorectal cancer. Colonic specimens were collected for histopathology, gene expression and flow cytometry analyses. Immune cell subsets in the mesenteric lymph nodes (MLN), spleen and colonic lamina propria lymphocytes (LPL) were phenotypically and functionally characterized. Mice treated with CLA or VSL#3 recovered faster from the acute inflammatory phase of disease and had lower disease severity in the chronic, tumor-bearing phase of disease. Adenoma and adenocarcinoma formation was also diminished by both treatments. VSL#3 increased the mRNA expression of TNF-α, angiostatin and PPAR γ whereas CLA decreased COX-2 levels. Moreover, VSL#3-treated mice had increased IL-17 expression in MLN CD4+ T cells and accumulation of Treg LPL and memory CD4+ T cells. Conclusions/Significance Both CLA and VSL#3 suppressed colon carcinogenesis, although VSL#3 showed greater anti-carcinogenic and anti-inflammatory activities than CLA. Mechanistically, CLA modulated expression of COX-2 levels in the colonic mucosa, whereas VSL#3 targeted regulatory mucosal CD4+ T cell responses.


Cellular Immunology | 2009

PPAR γ is highly expressed in F4/80hi adipose tissue macrophages and dampens adipose-tissue inflammation

Josep Bassaganya-Riera; Sarah Misyak; Amir J. Guri; Raquel Hontecillas

Macrophage infiltration into adipose tissue is a hallmark of obesity. We recently reported two phenotypically distinct subsets of adipose tissue macrophages (ATM) based on the surface expression of the glycoprotein F4/80 and responsiveness to treatment with a peroxisome proliferator-activated receptor (PPAR) gamma agonist. Hence, we hypothesized that F4/80(hi) and F4/80(lo) ATM differentially express PPAR gamma. This study phenotypically and functionally characterizes F4/80(hi) and F4/80(lo) ATM subsets during obesity. Changes in gene expression were also examined on sorted F4/80(lo) and F4/80(hi) ATM by quantitative real-time RT-PCR. We show that while F4/80(lo) macrophages predominate in adipose tissue of lean mice, obesity causes accumulation of both F4/80(lo) and F4/80(hi) ATM. Moreover, accumulation of F4/80(hi) ATM in adipose tissue is associated with impaired glucose tolerance. Phenotypically, F4/80(hi) ATM express greater amounts of CD11c, MHC II, CD49b, and CX3CR1 and produce more TNF-alpha, MCP-1, and IL-10 than F4/80(lo) ATM. Gene expression analyses of the sorted populations revealed that only the F4/80(lo) population produced IL-4, whereas the F4/80(hi) ATM expressed greater amounts of PPAR gamma, delta, CD36 and toll-like receptor-4. In addition, the deficiency of PPAR gamma in immune cells favors expression of M1 and impairs M2 macrophage marker expression in adipose tissue. Thus, PPAR gamma is differentially expressed in F4/80(hi) versus F4/80(low) ATM subsets and its deficiency favors a predominance of M1 markers in WAT.

Collaboration


Dive into the Raquel Hontecillas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Monica Viladomiu

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
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

Andrew Leber

Virginia Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar

Mireia Pedragosa

Virginia Bioinformatics Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge