Network


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

Hotspot


Dive into the research topics where Daniel Rodrigo-Torres is active.

Publication


Featured researches published by Daniel Rodrigo-Torres.


Gut | 2013

Transcriptome analysis identifies TNF superfamily receptors as potential therapeutic targets in alcoholic hepatitis

Silvia Affò; Marlene Dominguez; Juan José Lozano; Pau Sancho-Bru; Daniel Rodrigo-Torres; O. Morales-Ibanez; Montserrat Moreno; Cristina Millán; Aurora Loaeza-del-Castillo; José Altamirano; Juan Carlos García-Pagán; Vicente Arroyo; Pere Ginès; Juan Caballería; Robert F. Schwabe; Ramon Bataller

Objective Alcoholic hepatitis (AH) is a severe clinical condition that needs novel therapies. The identification of targets for therapy is hampered by the lack of animal models of advanced AH. The authors performed a translational study through a transcriptome analysis in patients with AH to identify new molecular targets. Design Hepatic gene expression profiling was assessed by DNA microarray in patients with AH (n=15) and normal livers (n=7). Functional analysis was assessed by gene set enrichment analysis. Quantitative PCR was performed in patients with AH (n=40), hepatitis C (n=18), non-alcoholic steatohepatitis (n=20) and in mouse models of acute and chronic liver injury. Protein expression was assessed by immunohistochemistry and western blotting. Results Gene expression analysis showed 207 genes >5-fold differentially expressed in patients with AH and revealed seven pathways differentially regulated including ‘cytokine–cytokine receptor interaction’. Several tumour necrosis factor (TNF) superfamily receptors, but not ligands, were overexpressed in AH. Importantly, Fn14 was the only TNF superfamily receptor exclusively upregulated in AH compared with other liver diseases and correlated with both 90-day mortality and severity of portal hypertension. Fn14 protein expression was detected in areas of fibrogenesis and in a population of hepatocytes. Fn14 expression was increased in experimental models of liver injury and was detected in progenitor cells. Conclusion Translational research revealed that TNF superfamily receptors are overexpressed in AH. Fn14, the receptor for TNF-like weak inducer of apoptosis, is selectively upregulated in patients with AH. TNF superfamily receptors could represent a potential target for therapy.


Hepatology | 2012

Liver progenitor cell markers correlate with liver damage and predict short-term mortality in patients with alcoholic hepatitis.

Pau Sancho-Bru; José Altamirano; Daniel Rodrigo-Torres; Mar Coll; Cristina Millán; Juan José Lozano; Rosa Miquel; Vicente Arroyo; Juan Caballería; Pere Ginès; Ramon Bataller

Alcoholic hepatitis (AH) is a severe condition developed in patients with underlying alcoholic liver disease. Ductular reaction has been associated with chronic alcohol consumption but there is no information regarding the extent of liver progenitor cell (LPC) proliferation in AH. The aim of this study was to investigate LPC markers in AH and its correlation with disease severity. Fifty‐nine patients with clinical and histological diagnosis of AH were included in the study. LPC markers were assessed by real‐time polymerase chain reaction (PCR) and immunohistochemistry. Standard logistic regression analysis and classification and regression trees (CART) analysis were used for statistical analysis. A microarray analysis showed an up‐regulation of LPC markers in patients with AH. Real‐time PCR demonstrated that epithelial cell adhesion molecule (EpCAM), Prominin‐1, and Keratin7 were significantly increased in patients with AH compared with normal livers (P ≤ 0.01), chronic hepatitis C (P ≤ 0.01), and HCV‐induced cirrhosis (P ≤ 0.01). Immunohistochemistry scores generated for Keratin7 and EpCAM demonstrated a good correlation with gene expression. Keratin7 gene expression correlated with liver failure as assessed by model for endstage liver disease score (r = 0.41, P = 0.006) and Maddreys discriminant function (r = 0.43, P = 0.004). Moreover, Keratin7 (OR1.14, P = 0.004) and Prominin‐1 (OR1.14, P = 0.002), but not EpCAM (OR1.16, P = 0.06), were identified as independent predictors of 90‐day mortality. CART analysis generated an algorithm based on the combination of Keratin7 and EpCAM gene expression that stratified three groups of patients with high, intermediate, and low short‐term mortality (89%, 33%, and 6%, respectively; area under the receiver operating curve 0.73, 95% confidence interval 0.60‐0.87). Keratin7 expression provided additional discrimination potential to the age, bilirubin, international normalization ratio, creatinine (ABIC) score. Conclusion: LPC markers correlate positively with severity of liver disease and short‐term mortality in AH patients. This study suggests that LPC proliferation may be an important feature of AH pathophysiology. (HEPATOLOGY 2012;55:1931–1941)


Hepatology | 2014

The biliary epithelium gives rise to liver progenitor cells

Daniel Rodrigo-Torres; Silvia Affò; Mar Coll; O. Morales-Ibanez; Cristina Millán; Delia Blaya; Anna Alvarez-Guaita; Carles Rentero; Juan José Lozano; Miguel Angel Maestro; Myriam Solar; Vicente Arroyo; Joan Caballería; Leo A. van Grunsven; Carlos Enrich; Pere Ginès; Ramon Bataller; P. Sancho-Bru

Severe liver diseases are characterized by expansion of liver progenitor cells (LPC), which correlates with disease severity. However, the origin and role of LPC in liver physiology and in hepatic injury remains a contentious topic. We found that ductular reaction cells in human cirrhotic livers express hepatocyte nuclear factor 1 homeobox B (HNF1β). However, HNF1β expression was not present in newly generated epithelial cell adhesion molecule (EpCAM)‐positive hepatocytes. In order to investigate the role of HNF1β‐expressing cells we used a tamoxifen‐inducible Hnf1βCreER/R26RYfp/LacZ mouse to lineage‐trace Hnf1β+ biliary duct cells and to assess their contribution to LPC expansion and hepatocyte generation. Lineage tracing demonstrated no contribution of HNF1β+ cells to hepatocytes during liver homeostasis in healthy mice or after loss of liver mass. After acute acetaminophen or carbon tetrachloride injury no contribution of HNF1β+ cells to hepatocyte was detected. We next assessed the contribution of Hnf1β+‐derived cells following two liver injury models with LPC expansion, a diethoxycarbonyl‐1,4‐dihydro‐collidin (DDC)‐diet and a choline‐deficient ethionine‐supplemented (CDE)‐diet. The contribution of Hnf1β+ cells to liver regeneration was dependent on the liver injury model. While no contribution was observed after DDC‐diet treatment, mice fed with a CDE‐diet showed a small population of hepatocytes derived from Hnf1β+ cells that were expanded to 1.86% of total hepatocytes after injury recovery. Genome‐wide expression profile of Hnf1β+‐derived cells from the DDC and CDE models indicated that no contribution of LPC to hepatocytes was associated with LPC expression of genes related to telomere maintenance, inflammation, and chemokine signaling pathways. Conclusion: HNF1β+ biliary duct cells are the origin of LPC. HNF1β+ cells do not contribute to hepatocyte turnover in the healthy liver, but after certain liver injury, they can differentiate to hepatocytes contributing to liver regeneration. (Hepatology 2014;60:1367–1377)


Gut | 2014

CCL20 mediates lipopolysaccharide induced liver injury and is a potential driver of inflammation and fibrosis in alcoholic hepatitis

Silvia Affò; O. Morales-Ibanez; Daniel Rodrigo-Torres; José Altamirano; Delia Blaya; Cristina Millán; Mar Coll; Jorge Matias Caviglia; Vicente Arroyo; Juan Caballería; Robert F. Schwabe; Pere Ginès; Ramon Bataller; P. Sancho-Bru

Objective Chemokines are known to play an important role in the pathophysiology of alcoholic hepatitis (AH), a form of acute-on-chronic liver injury frequently mediated by gut derived lipopolysaccharide (LPS). In our study, we hypothesise that chemokine CCL20, one of the most upregulated chemokines in patients with AH, is implicated in the pathogenesis of AH by mediating LPS induced liver injury. Design CCL20 gene expression and serum levels and their correlation with disease severity were assessed in patients with AH. Cellular sources of CCL20 and its biological effects were evaluated in vitro and in vivo in chronic, acute and acute-on-chronic experimental models of carbon tetrachloride and LPS induced liver injury. RNA interference technology was used to knockdown CCL20 in vivo. Results CCL20 hepatic and serum levels were increased in patients with AH and correlated with the degree of fibrosis, portal hypertension, endotoxaemia, disease severity scores and short term mortality. Moreover, CCL20 expression was increased in animal models of liver injury and particularly under acute-on-chronic conditions. Macrophages and hepatic stellate cells (HSCs) were identified as the main CCL20 producing cell types. Silencing CCL20 in vivo reduced LPS induced aspartate aminotransferase and lactate dehydrogenase serum levels and hepatic proinflammatory and profibrogenic genes. CCL20 induced proinflammatory and profibrogenic effects in cultured primary HSCs. Conclusions Our results suggest that CCL20 upregulation is strongly associated with LPS and may not only represent a new potential biomarker to predict outcome in patients with AH but also an important mediator linking hepatic inflammation, injury and fibrosis in AH.


Scientific Reports | 2015

Integrative miRNA and Gene Expression Profiling Analysis of Human Quiescent Hepatic Stellate Cells

Mar Coll; Adil El Taghdouini; L. Perea; Inge Mannaerts; Maria Vila-Casadesús; Delia Blaya; Daniel Rodrigo-Torres; Silvia Affò; O. Morales-Ibanez; Isabel Graupera; Juan José Lozano; Mustapha Najimi; Etienne Sokal; Joeri Lambrecht; Pere Ginès; Leo A. van Grunsven; Pau Sancho-Bru

Unveiling the regulatory pathways maintaining hepatic stellate cells (HSC) in a quiescent (q) phenotype is essential to develop new therapeutic strategies to treat fibrogenic diseases. To uncover the miRNA-mRNA regulatory interactions in qHSCs, HSCs were FACS-sorted from healthy livers and activated HSCs (aHSCs) were generated in vitro. MiRNA Taqman array analysis showed HSCs expressed a low number of miRNAs (n = 259), from which 47 were down-regulated and 212 up-regulated upon activation. Computational integration of miRNA and gene expression profiles revealed that 66% of qHSC-associated miRNAs correlated with more than 6 altered target mRNAs (17,28 ± 10,7 targets/miRNA) whereas aHSC-associated miRNAs had an average of 1,49 targeted genes. Interestingly, interaction networks generated by miRNA-targeted genes in qHSCs were associated with key HSC activation processes. Next, selected miRNAs were validated in healthy and cirrhotic human livers and miR-192 was chosen for functional analysis. Down-regulation of miR-192 in HSCs was found to be an early event during fibrosis progression in mouse models of liver injury. Moreover, mimic assays for miR-192 in HSCs revealed its role in HSC activation, proliferation and migration. Together, these results uncover the importance of miRNAs in the maintenance of the qHSC phenotype and form the basis for understanding the regulatory networks in HSCs.


Gut | 2016

Integrative microRNA profiling in alcoholic hepatitis reveals a role for microRNA-182 in liver injury and inflammation

Delia Blaya; Mar Coll; Daniel Rodrigo-Torres; Maria Vila-Casadesús; José Altamirano; M. Llopis; Isabel Graupera; L. Perea; Beatriz Aguilar-Bravo; Alba Díaz; Jesus M. Banales; Joan Clària; Juan José Lozano; Ramon Bataller; Juan Caballería; Pere Ginès; Pau Sancho-Bru

Objective MicroRNAs (miRNAs) are well-known regulators of disease pathogenesis and have great potential as biomarkers and therapeutic targets. We aimed at profiling miRNAs in alcoholic hepatitis (AH) and identifying miRNAs potentially involved in liver injury. Design MiRNA profiling was performed in liver samples from patients with AH, alcohol liver disease, non-alcoholic steatohepatitis, HCV disease and normal liver tissue. Expression of miRNAs was assessed in liver and serum from patients with AH and animal models. Mimic and decoy miR-182 were used in vitro and in vivo to evaluate miR-182s biological functions. Results MiRNA expression profile in liver was highly altered in AH and distinctive from alcohol-induced cirrhotic livers. Moreover, we identified a set of 18 miRNAs predominantly expressed in AH as compared with other chronic liver conditions. Integrative miRNA-mRNA functional analysis revealed the association of AH-altered miRNAs with nuclear receptors, IGF-1 signalling and cholestasis. Interestingly, miR-182 was the most highly expressed miRNA in AH, which correlated with degree of ductular reaction, disease severity and short-term mortality. MiR-182 mimic induced an upregulation of inflammatory mediators in biliary cells. At experimental level, miR-182 was increased in biliary cells in mice fed with 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC) diet but not upregulated by alcohol intake or fibrosis. Inhibition of miR-182 in DDC-fed mice reduced liver damage, bile acid accumulation and inflammatory response. Conclusions AH is characterised by a deregulated miRNA profile, including miR-182, which is associated with disease severity and liver injury. These results highlight the potential of miRNAs as therapeutic targets and biomarkers in AH.


Scientific Reports | 2016

LPS-TLR4 Pathway Mediates Ductular Cell Expansion in Alcoholic Hepatitis

Gemma Odena; Jiegen Chen; Juan José Lozano; José Altamirano; Daniel Rodrigo-Torres; Silvia Affò; O. Morales-Ibanez; Hiroshi Matsushita; Jian Zou; Raluca Dumitru; Juan Caballería; Pere Ginès; Vicente Arroyo; Min You; Pierre-Emmanuel Rautou; Dominique Valla; Fulton T. Crews; Ekihiro Seki; Pau Sancho-Bru; Ramon Bataller

Alcoholic hepatitis (AH) is the most severe form of alcoholic liver disease for which there are no effective therapies. Patients with AH show impaired hepatocyte proliferation, expansion of inefficient ductular cells and high lipopolysaccharide (LPS) levels. It is unknown whether LPS mediates ductular cell expansion. We performed transcriptome studies and identified keratin 23 (KRT23) as a new ductular cell marker. KRT23 expression correlated with mortality and LPS serum levels. LPS-TLR4 pathway role in ductular cell expansion was assessed in human and mouse progenitor cells, liver slices and liver injured TLR4 KO mice. In AH patients, ductular cell expansion correlated with portal hypertension and collagen expression. Functional studies in ductular cells showed that KRT23 regulates collagen expression. These results support a role for LPS-TLR4 pathway in promoting ductular reaction in AH. Maneuvers aimed at decreasing LPS serum levels in AH patients could have beneficial effects by preventing ductular reaction development.


Hepatology | 2017

Pentraxin‐3 modulates lipopolysaccharide‐induced inflammatory response and attenuates liver injury

L. Perea; Mar Coll; Lucía Sanjurjo; Delia Blaya; Adil El Taghdouini; Daniel Rodrigo-Torres; José Altamirano; Isabel Graupera; Beatriz Aguilar-Bravo; M. Llopis; Julia Vallverdú; Joan Caballería; Leo A. van Grunsven; Maria-Rosa Sarrias; Pere Ginès; Pau Sancho-Bru

Acute‐on‐chronic liver injury is characterized by an important inflammatory response frequently associated with endotoxemia. In this context, acute‐phase proteins such as Pentraxin‐3 (PTX3) are released; however, little is known about their role in chronic liver disease. The aim of this study was to elucidate the role of PTX3 in liver injury. The role of PTX3 was evaluated in cultured human cells, liver tissue slices, and mice with acute‐on‐chronic liver injury. PTX3 expression was assessed in tissue and serum samples from 54 patients with alcoholic hepatitis. PTX3 expression was up‐regulated in animal models of liver injury and strongly induced by lipopolysaccharide (LPS). Liver cell fractionation showed that macrophages and activated hepatic stellate cells were the main cell types expressing PTX3 in liver injury. Ex vivo and in vivo studies showed that PTX3 treatment attenuated LPS‐induced liver injury, inflammation, and cell recruitment. Mechanistically, PTX3 mediated the hepatic stellate cell wound‐healing response. Moreover, PTX3 modulated LPS‐induced inflammation in human primary liver macrophages and peripheral monocytes by enhancing a TIR domain–containing adapter‐inducing interferon–dependent response and favoring a macrophage interleukin‐10‐like phenotype. Additionally, hepatic and plasma PTX3 levels were increased in patients with alcoholic hepatitis, a prototypic acute‐on‐chronic condition; and its expression correlated with disease severity scores, endotoxemia, infections, and short‐term mortality, thus suggesting that expression of PTX3 found in patients could be a counterregulatory response to injury. Conclusion: Experimental and human evidence suggests that, in addition to being a potential biomarker for alcoholic hepatitis, PTX3 participates in the wound‐healing response and attenuates LPS‐induced liver injury and inflammation; therefore, administration of PTX3 could be a promising therapeutic strategy in acute‐on‐chronic conditions, particularly those associated with endotoxemia. (Hepatology 2017;66:953–968).


Hepatology | 2017

Pentraxin‐3 Modulates LPS‐induced Inflammatory Response and Attenuates Liver Injury

L. Perea; Mar Coll; Lucía Sanjurjo; Delia Blaya; Adil El Taghdouini; Daniel Rodrigo-Torres; José Altamirano; Isabel Graupera; Beatriz Aguilar-Bravo; M. Llopis; Julia Vallverdú; Joan Caballería; Leo A. van Grunsven; Maria-Rosa Sarrias; Pere Ginès; Pau Sancho-Bru

Acute‐on‐chronic liver injury is characterized by an important inflammatory response frequently associated with endotoxemia. In this context, acute‐phase proteins such as Pentraxin‐3 (PTX3) are released; however, little is known about their role in chronic liver disease. The aim of this study was to elucidate the role of PTX3 in liver injury. The role of PTX3 was evaluated in cultured human cells, liver tissue slices, and mice with acute‐on‐chronic liver injury. PTX3 expression was assessed in tissue and serum samples from 54 patients with alcoholic hepatitis. PTX3 expression was up‐regulated in animal models of liver injury and strongly induced by lipopolysaccharide (LPS). Liver cell fractionation showed that macrophages and activated hepatic stellate cells were the main cell types expressing PTX3 in liver injury. Ex vivo and in vivo studies showed that PTX3 treatment attenuated LPS‐induced liver injury, inflammation, and cell recruitment. Mechanistically, PTX3 mediated the hepatic stellate cell wound‐healing response. Moreover, PTX3 modulated LPS‐induced inflammation in human primary liver macrophages and peripheral monocytes by enhancing a TIR domain–containing adapter‐inducing interferon–dependent response and favoring a macrophage interleukin‐10‐like phenotype. Additionally, hepatic and plasma PTX3 levels were increased in patients with alcoholic hepatitis, a prototypic acute‐on‐chronic condition; and its expression correlated with disease severity scores, endotoxemia, infections, and short‐term mortality, thus suggesting that expression of PTX3 found in patients could be a counterregulatory response to injury. Conclusion: Experimental and human evidence suggests that, in addition to being a potential biomarker for alcoholic hepatitis, PTX3 participates in the wound‐healing response and attenuates LPS‐induced liver injury and inflammation; therefore, administration of PTX3 could be a promising therapeutic strategy in acute‐on‐chronic conditions, particularly those associated with endotoxemia. (Hepatology 2017;66:953–968).


PLOS ONE | 2015

Chemokine Receptor Ccr6 Deficiency Alters Hepatic Inflammatory Cell Recruitment and Promotes Liver Inflammation and Fibrosis

Silvia Affò; Daniel Rodrigo-Torres; Delia Blaya; O. Morales-Ibanez; Mar Coll; Cristina Millán; José Altamirano; Vicente Arroyo; Joan Caballería; Ramon Bataller; Pere Ginès; Pau Sancho-Bru

Chronic liver diseases are characterized by a sustained inflammatory response in which chemokines and chemokine-receptors orchestrate inflammatory cell recruitment. In this study we investigated the role of the chemokine receptor CCR6 in acute and chronic liver injury. In the absence of liver injury Ccr6 -/- mice presented a higher number of hepatic macrophages and increased expression of pro-inflammatory cytokines and M1 markers Tnf-α, Il6 and Mcp1. Inflammation and cell recruitment were increased after carbon tetrachloride-induced acute liver injury in Ccr6 -/- mice. Moreover, chronic liver injury by carbon tetrachloride in Ccr6 -/- mice was associated with enhanced inflammation and fibrosis, altered macrophage recruitment, enhanced CD4+ cells and a reduction in Th17 (CD4+IL17+) and mature dendritic (MHCII+CD11c+) cells recruitment. Clodronate depletion of macrophages in Ccr6 -/- mice resulted in a reduction of hepatic pro-inflammatory and pro-fibrogenic markers in the absence and after liver injury. Finally, increased CCR6 hepatic expression in patients with alcoholic hepatitis was found to correlate with liver expression of CCL20 and severity of liver disease. In conclusion, CCR6 deficiency affects hepatic inflammatory cell recruitment resulting in the promotion of hepatic inflammation and fibrosis.

Collaboration


Dive into the Daniel Rodrigo-Torres's collaboration.

Top Co-Authors

Avatar

Mar Coll

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Delia Blaya

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Pere Ginès

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

L. Perea

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge