Network


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

Hotspot


Dive into the research topics where Luis Enrique Gómez-Quiroz is active.

Publication


Featured researches published by Luis Enrique Gómez-Quiroz.


Cancer Research | 2007

Loss of Hepatocyte Growth Factor/c-Met Signaling Pathway Accelerates Early Stages of N-nitrosodiethylamine-Induced Hepatocarcinogenesis

Taro Takami; Pal Kaposi-Novak; Koichi Uchida; Luis Enrique Gómez-Quiroz; Elizabeth A. Conner; Valentina M. Factor; Snorri S. Thorgeirsson

Hepatocyte growth factor (HGF) has been reported to have both positive and negative effects on carcinogenesis. Here, we show that the loss of c-Met signaling in hepatocytes enhanced rather than suppressed the early stages of chemical hepatocarcinogenesis. c-Met conditional knockout mice (c-metfl/fl, AlbCre+/-; MetLivKO) treated with N-nitrosodiethylamine developed significantly more and bigger tumors and with a shorter latency compared with control (w/w, AlbCre+/-; Cre-Ctrl) mice. Accelerated tumor development was associated with increased rate of cell proliferation and prolonged activation of epidermal growth factor receptor (EGFR) signaling. MetLivKO livers treated with N-nitrosodiethylamine also displayed elevated lipid peroxidation, decreased ratio of reduced glutathione to oxidized glutathione, and up-regulation of superoxide dismutase 1 and heat shock protein 70, all consistent with increased oxidative stress. Likewise, gene expression profiling done at 3 and 5 months after N-nitrosodiethylamine treatment revealed up-regulation of genes associated with cell proliferation and stress responses in c-Met mutant livers. The negative effects of c-Met deficiency were reversed by chronic p.o. administration of antioxidant N-acetyl-L-cysteine. N-acetyl-L-cysteine blocked the EGFR activation and reduced the N-nitrosodiethylamine-initiated hepatocarcinogenesis to the levels of Cre-Ctrl mice. These results argue that intact HGF/c-Met signaling is essential for maintaining normal redox homeostasis in the liver and has tumor suppressor effect(s) during the early stages of N-nitrosodiethylamine-induced hepatocarcinogenesis.


Journal of Biological Chemistry | 2008

Hepatocyte-specific c-Met Deletion Disrupts Redox Homeostasis and Sensitizes to Fas-mediated Apoptosis

Luis Enrique Gómez-Quiroz; Valentina M. Factor; Pal Kaposi-Novak; Cédric Coulouarn; Elizabeth A. Conner; Snorri S. Thorgeirsson

The hepatocyte growth factor and its receptor c-Met direct a pleiotropic signal transduction pathway that controls cell survival. We previously demonstrated that mice lacking c-Met (Met-KO) in hepatocytes were hypersensitive to Fas-induced liver injury. In this study, we used primary hepatocytes isolated from Met-KO and control (Cre-Ctrl) mice to address more directly the protective effects of c-Met signaling. Loss of c-Met function increased sensitivity to Fas-mediated apoptosis. Hepatocyte growth factor suppressed apoptosis in Cre-Ctrl but not Met-KO hepatocytes concurrently with up-regulation of NF-κB and major antiapoptotic proteins Bcl-2 and Bcl-xL. Intriguingly, Met-KO hepatocytes exhibited intrinsic activation of NF-κBas well as Bcl-2 and Bcl-xL. Furthermore, unchallenged Met-KO cells displayed oxidative stress as evidenced by overproduction of reactive oxygen species, which was associated with greater NADPH and Rac1 activities, was blocked by the known NADPH oxidase inhibitors, and was paralleled by increased lipid peroxidation and reduced glutathione (GSH) content. N-Acetylcysteine, an antioxidant and GSH precursor, significantly reduced Jo2-induced cell death. Conversely, the GSH-depleting agent buthionine sulfoximine completely abolished the protective effects of N-acetylcysteine in Met-KO hepatocytes. In conclusion, genetic inactivation of c-Met in mouse hepatocytes caused defects in redox regulation, which may account for the increased sensitivity to Fas-induced apoptosis and adaptive up-regulation of NF-κB survival signaling. These data provide evidence that intact c-Met signaling is a critical factor in the protection against excessive generation of endogenous reactive oxygen species.


Journal of Hepatology | 2015

Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer

Jens U. Marquardt; Luis Enrique Gómez-Quiroz; Lucrecia O. Arreguin Camacho; Federico Pinna; Yun-Han Lee; Mitsuteru Kitade; Mayrel Palestino Domínguez; D Castven; Kai Breuhahn; Elizabeth A. Conner; Peter R. Galle; Jesper B. Andersen; Valentina M. Factor; Snorri S. Thorgeirsson

BACKGROUND & AIMS The cancer stem cells (CSCs) have important therapeutic implications for multi-resistant cancers including hepatocellular carcinoma (HCC). Among the key pathways frequently activated in liver CSCs is NF-κB signaling. METHODS We evaluated the CSCs-depleting potential of NF-κB inhibition in liver cancer achieved by the IKK inhibitor curcumin, RNAi and specific peptide SN50. The effects on CSCs were assessed by analysis of side population (SP), sphere formation and tumorigenicity. Molecular changes were determined by RT-qPCR, global gene expression microarray, EMSA, and Western blotting. RESULTS HCC cell lines exposed to curcumin exhibited differential responses to curcumin and were classified as sensitive and resistant. In sensitive lines, curcumin-mediated induction of cell death was directly related to the extent of NF-κB inhibition. The treatment also led to a selective CSC-depletion as evidenced by a reduced SP size, decreased sphere formation, down-regulation of CSC markers and suppressed tumorigenicity. Similarly, NF-κB inhibition by SN50 and siRNA against p65 suppressed tumor cell growth. In contrast, curcumin-resistant cells displayed a paradoxical increase in proliferation and expression of CSC markers. Mechanistically, an important component of the CSC-depleting activity of curcumin could be attributed to a NF-κB-mediated HDAC inhibition. Co-administration of the class I/II HDAC inhibitor trichostatine sensitized resistant cells to curcumin. Further, integration of a predictive signature of curcumin sensitivity with human HCC database indicated that HCCs with poor prognosis and progenitor features are most likely to benefit from NF-κB inhibition. CONCLUSIONS These results demonstrate that blocking NF-κB can specifically target CSC populations and suggest a potential for combined inhibition of NF-κB and HDAC signaling for treatment of liver cancer patients with poor prognosis.


Cell Biology and Toxicology | 2009

Acetaldehyde-induced mitochondrial dysfunction sensitizes hepatocytes to oxidative damage

Blanca Eugenia Farfán Labonne; Mario Gutiérrez; Luis Enrique Gómez-Quiroz; Mina Konigsberg Fainstein; Verónica Souza; Oscar Flores Flores; Victor Ortíz; Elizabeth Hernández; David Kershenobich; María Concepción Gutiérrez-Ruiz

Acetaldehyde (Ac), the main metabolite of ethanol oxidation, is a very reactive compound involved in alcohol-induced liver damage. In the present work, we studied the effect of Ac in mitochondria functionality. Mitochondria from Wistar rats were isolated and treated with Ac. Ac decreased respiratory control by 50% which was associated with a decrease in adenosine triphosphate content (28.5%). These results suggested that Ac could be inducing changes in cell redox status. We determined protein oxidation, superoxide dismutase (SOD) activity, and glutathione ratio, indicating that Ac induced an enhanced oxidation of proteins and a decrease in SOD activity (90%) and glutathione/oxidized GSH ratio (36%). The data suggested that Ac-induced oxidative stress mediated by mitochondria dysfunction can lead to cell sensitization and to a second oxidative challenge. We pretreated hepatocytes with Ac followed by treatment with antimycin A, and this experiment revealed a noticeable decrease in cell viability, determined by neutral red assay, in comparison with cells treated with Ac alone. Our data demonstrate that Ac impairs mitochondria functionality generating oxidative stress that sensitizes cells to a second damaging signal contributing to the development of alcoholic liver disease.


Free Radical Biology and Medicine | 2009

Hepatocyte growth factor protects hepatocytes against oxidative injury induced by ethanol metabolism.

Argelia Valdés-Arzate; Armando Luna; Cynthia Licona; Dahn L. Clemens; Verónica Souza; Elizabeth Hernández; David Kershenobich; María Concepción Gutiérrez-Ruiz; Luis Enrique Gómez-Quiroz

Hepatocyte growth factor (HGF) is involved in many cellular responses, such as mitogenesis and apoptosis protection; however, its effect against oxidative injury induced by ethanol metabolism is not well understood. The aim of this work was to address the mechanism of HGF-induced protection against ethanol-generated oxidative stress damage in the human cell line VL-17A (cytochrome P450 2E1/alcohol dehydrogenase-transfected HepG2 cells). Cells were pretreated with 50 ng/ml HGF for 12 h and then treated with 100 mM ethanol for 0-48 h. Some parameters of oxidative damage were evaluated. We found that ethanol induced peroxide formation (3.3-fold) and oxidative damage as judged by lipid peroxidation (5.4-fold). Damage was prevented by HGF. To address the mechanisms of HGF-induced protection we investigated the cellular antioxidant system. We found that HGF increased the GSH/GSSG ratio, as well as SOD1, catalase, and gamma-glutamylcysteine synthetase expression. To explore the signaling pathways involved in this process, VL-17A cells were pretreated with inhibitors against PI3K, Akt, and NF-kappaB. We found that all treatments decreased the expression of the antioxidant enzymes, thus abrogating the HGF-induced protection against oxidative stress. Our results demonstrate that HGF protects cells from the oxidative damage induced by ethanol metabolism by a mechanism driven by NF-kappaB and PI3K/Akt signaling.


Toxicology Letters | 2009

NADPH oxidase and ERK1/2 are involved in cadmium induced-STAT3 activation in HepG2 cells

Verónica Souza; Ma. del Carmen Escobar; Elizabeth Hernández; Luis Enrique Gómez-Quiroz; Ma. Concepción Gutiérrez Ruiz

The molecular mechanism of Cd-induced signal transduction is not well understood. The aims of this study were to determine the system that generates reactive oxygen species in response to Cd that contribute to intracellular signaling on the activation of the STAT3 pathway in HepG2 cells and to address the participation of STAT3 in the production of Hsp70. Cadmium induced a significant increase in STAT3 DNA-binding after 1h treatment. Serine phosphorylation of STAT3 was observed as a result of cadmium treatment while no tyrosine phosphorylation was detected. Cells were pretreated with inhibitors for several ROS generating systems, only diphenylen iodonium, an inhibitor of NADPH oxidase, decreased STAT3 activation. Cd induced 2.6-fold NADPH oxidase activity. Antioxidant treatment with pegylated-catalase reduced STAT3 activation. Cells were pretreated with different MAPKs inhibitors. ERK contributes in approximately 60%, and JNK in a small proportion, while p38 does not contribute in STAT3 activation. Cells were pretreated with a specific STAT3 peptide inhibitor that decreased the Cd-induced Hsp70 expression. Data suggest that STAT3 is phosphorylated at serine 727 by a Cd stress-activated signaling pathway inducing NADPH oxidase activity which produced ROS, leading ERK activation. MAPK promotes STAT3 phosphorylation that could induce a protective mechanism against Cd toxicity.


Toxicological Sciences | 2013

Hepatocyte Growth Factor Protects Against Isoniazid/Rifampicin-Induced Oxidative Liver Damage

Cristina Enríquez-Cortina; Maylin Almonte-Becerril; Denise Clavijo-Cornejo; Mayrel Palestino-Domínguez; Oscar Bello-Monroy; Natalia Nuño; Anayelly López; Verónica Souza; Rogelio Hernández-Pando; Linda Muñoz; María Concepción Gutiérrez-Ruiz; Luis Enrique Gómez-Quiroz

The worldwide increment of multidrug- and extensively drug-resistant tuberculosis has emphasized the importance of looking for new options in therapeutics. Long-time usage or higher doses of isoniazid and rifampicin have been considered for the treatment of multidrug-resistant tuberculosis; however, the risk of liver failure is proportionally increased. Hepatocyte growth factor (HGF) is a multitask growth factor that stimulates both antiapoptotic and antioxidant responses that counteract the toxic effects of drug metabolism in the liver. The present work was focused to address the antioxidant and antiapoptotic effects of HGF on isoniazid- and rifampicin-induced hepatotoxicity. BALB/c mice were subjected to rifampicin (150mg/kg, intragavage [ig]) plus isoniazid (75mg/kg, ig) for 7 days. Increments in alanine aminotransferase activity, steatosis, apoptosis, and oxidative stress markers were found in animals. Recombinant HGF (iv) prevented all the harmful effects by increasing the activation of Erk1/2 and PKCδ signaling pathways and glutathione (GSH) synthesis. Furthermore, inhibition of endogenous HGF with anti-HGF antibody (iv) enhanced the isoniazid- and rifampicin-induced oxidative stress damage and decreased the GSH content, aggravating liver damage. In conclusion, HGF demonstrated to be a good protective factor against antituberculosis drug-induced hepatotoxicity and could be considered a good adjuvant factor for the use of high doses of or the reintroduction of these antituberculosis drugs.


Biochimica et Biophysica Acta | 2012

Loss of c-Met Accelerates Development of Liver Fibrosis in Response to CCl4 Exposure through Deregulation of Multiple Molecular Pathways

Jens U. Marquardt; Daekwan Seo; Luis Enrique Gómez-Quiroz; Koichi Uchida; Matthew C. Gillen; Mitsuteru Kitade; Pal Kaposi-Novak; Elizabeth A. Conner; Valentina M. Factor; Snorri S. Thorgeirsson

HGF/c-Met signaling plays a pivotal role in hepatocyte survival and tissue remodeling during liver regeneration. HGF treatment accelerates resolution of fibrosis in experimental animal models. Here, we utilized Met(fl/fl);Alb-Cre(+/-) conditional knockout mice and a carbon tetrachloride(CCl(4))-induced liver fibrosis model to formally address the role of c-Met signaling in hepatocytes in the context of chronic tissue injury. Histological changes during injury (4weeks) and healing phase (4weeks) were monitored by immunohistochemistry; expression levels of selected key fibrotic molecules were evaluated by western blotting, and time-dependent global transcriptomic changes were examined using a microarray platform. Loss of hepatocyte c-Met signaling altered hepatic microenvironment and aggravated hepatic fibrogenesis. Greater liver damage was associated with decreased hepatocyte proliferation, excessive stellate cell activation and rapid dystrophic calcification of necrotic areas. Global transcriptome analysis revealed a broad impact of c-Met on critical signaling pathways associated with fibrosis. Loss of hepatocyte c-Met caused a strong deregulation of chemotactic and inflammatory signaling (MCP-1, RANTES, Cxcl10) in addition to modulation of genes involved in reorganization of the cytoskeletal network (Actb, Tuba1a, Tuba8), intercellular communications and adhesion (Adam8, Icam1, Itgb2), control of cell proliferation (Ccng2, Csnk2a, Cdc6, cdk10), DNA damage and stress response (Rad9, Rad52, Ercc4, Gsta1 and 2, Jun). Our study demonstrates that deletion of c-Met receptor in hepatocytes results in pronounced changes in hepatic metabolism and microenvironment, and establishes an essential role for c-Met in maintaining the structural integrity and adaptive plasticity of the liver under adverse conditions.


Liver International | 2007

Liver fibrosis: searching for cell model answers

Ma.Concepción Gutiérrez-Ruiz; Luis Enrique Gómez-Quiroz

Hepatic stellate cells (HSC) are the principal fibrogenic cell type in the liver. Progress in understanding the cellular and molecular basis for the development and progression of liver fibrosis could be possible by the development of methods to isolate HSC from rodents and human liver. Growth of stellate cells on plastic led to a phenotypic response known as activation, which paralleled closely the response of these cells to injury in vivo. Actually, much of the current knowledge of stellate cell behaviour has been gained through primary culture studies, particularly from rats. Also, different laboratories that have established hepatic stellate cell lines from rats and humans have provided a stable and unlimited source of cells that express specific functions, making them suitable for culture‐based studies of hepatic fibrosis. From these in vitro models grew a large body of information characterizing stellate cell activation, cytokine signalling, intracellular pathways regulating liver fibrogenesis, production of extracellular matrix proteins and development of antifibrotic drugs.


Cancer Research | 2014

Antitumor effects in hepatocarcinoma of isoform-selective inhibition of HDAC2

Yun-Han Lee; Daekwan Seo; Kyung Ju Choi; Jesper B. Andersen; Min Ah Won; Mitsuteru Kitade; Luis Enrique Gómez-Quiroz; Adam Judge; Jens U. Marquardt; Chiara Raggi; Elizabeth A. Conner; Ian Maclachlan; Valentina M. Factor; Snorri S. Thorgeirsson

Histone deacetylase 2 (HDAC2) is a chromatin modifier involved in epigenetic regulation of cell cycle, apoptosis, and differentiation that is upregulated commonly in human hepatocellular carcinoma (HCC). In this study, we show that specific targeting of this HDAC isoform is sufficient to inhibit HCC progression. siRNA-mediated silencing of HDAC inhibited HCC cell growth by blocking cell-cycle progression and inducing apoptosis. These effects were associated with deregulation of HDAC-regulated genes that control cell cycle, apoptosis, and lipid metabolism, specifically, by upregulation of p27 and acetylated p53 and by downregulation of CDK6 and BCL2. We found that HDAC2 silencing in HCC cells also strongly inhibited PPARγ signaling and other regulators of glycolysis (ChREBPα and GLUT4) and lipogenesis (SREBP1C and FAS), eliciting a marked decrease in fat accumulation. Notably, systemic delivery of HDAC2 siRNA encapsulated in lipid nanoparticles was sufficient to blunt the growth of human HCC in a murine xenograft model. Our findings offer preclinical proof-of-concept for HDAC2 blockade as a systemic therapy for liver cancer.

Collaboration


Dive into the Luis Enrique Gómez-Quiroz's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Verónica Souza

Universidad Autónoma Metropolitana

View shared research outputs
Top Co-Authors

Avatar

Elizabeth Hernández

Universidad Autónoma Metropolitana

View shared research outputs
Top Co-Authors

Avatar

Roxana U. Miranda

Universidad Autónoma Metropolitana

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Kershenobich

National Autonomous University of Mexico

View shared research outputs
Top Co-Authors

Avatar

Ma.Concepción Gutiérrez-Ruiz

Universidad Autónoma Metropolitana

View shared research outputs
Top Co-Authors

Avatar

Denise Clavijo-Cornejo

Universidad Autónoma Metropolitana

View shared research outputs
Top Co-Authors

Avatar

Elizabeth A. Conner

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge