Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
1.
J. physiol. biochem ; 79(4): 881-890, nov. 2023. graf, ilus
Article in English | IBECS | ID: ibc-227559

ABSTRACT

Ocoxin is a nutritional supplement that has been shown to exert antioxidant and immunomodulatory responses in patients with chronic hepatitis C. The present work aimed to determine the effects of Ocoxin on activated hepatic stellate cells (HSC), the cell type mainly responsible for collagen deposition in the fibrotic liver. Ocoxin was found to reduce the survival of a cell line of immortalized non-tumoral rat HSC in a dose–response fashion and to diminish collagen type I levels. This latter effect was observed even at doses not affecting cell survival, pointing to an antifibrogenic action for the supplement. The decrease in viability exerted by Ocoxin on HSC correlated with an increase in histone-associated fragments in the cytoplasm and with increased activity of caspase-3, indicating the induction of apoptosis. To determine the molecular mechanisms mediating Ocoxin-induced apoptosis, the activation of members of the MAPK family was analyzed. Incubation of HSC with Ocoxin caused a transient and dramatic enhancement on ERK, JNK, and p38 MAPK phosphorylation levels. Using specific inhibitors for these enzymes, p38 MAPK was identified as a key mediator of the apoptotic effect of Ocoxin on HSC. (AU)


Subject(s)
Animals , Rats , Hepatic Stellate Cells/metabolism , Plant Extracts/metabolism , Apoptosis , Cells, Cultured , Liver Cirrhosis/metabolism , Phosphorylation , p38 Mitogen-Activated Protein Kinases/metabolism
2.
J Physiol Biochem ; 79(4): 881-890, 2023 Nov.
Article in English | MEDLINE | ID: mdl-35239161

ABSTRACT

Ocoxin is a nutritional supplement that has been shown to exert antioxidant and immunomodulatory responses in patients with chronic hepatitis C. The present work aimed to determine the effects of Ocoxin on activated hepatic stellate cells (HSC), the cell type mainly responsible for collagen deposition in the fibrotic liver. Ocoxin was found to reduce the survival of a cell line of immortalized non-tumoral rat HSC in a dose-response fashion and to diminish collagen type I levels. This latter effect was observed even at doses not affecting cell survival, pointing to an antifibrogenic action for the supplement. The decrease in viability exerted by Ocoxin on HSC correlated with an increase in histone-associated fragments in the cytoplasm and with increased activity of caspase-3, indicating the induction of apoptosis. To determine the molecular mechanisms mediating Ocoxin-induced apoptosis, the activation of members of the MAPK family was analyzed. Incubation of HSC with Ocoxin caused a transient and dramatic enhancement on ERK, JNK, and p38 MAPK phosphorylation levels. Using specific inhibitors for these enzymes, p38 MAPK was identified as a key mediator of the apoptotic effect of Ocoxin on HSC.


Subject(s)
Hepatic Stellate Cells , Plant Extracts , Rats , Humans , Animals , Hepatic Stellate Cells/metabolism , Phosphorylation , Plant Extracts/metabolism , Liver Cirrhosis/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cells, Cultured , Apoptosis
3.
Int Rev Cell Mol Biol ; 372: 55-96, 2022.
Article in English | MEDLINE | ID: mdl-36064267

ABSTRACT

Inborn errors of metabolism (IEM) encompass a group of monogenic diseases affecting both pediatric and adult populations and currently lack effective treatments. Some IEM such as familial hypercholesterolemia or X-linked protoporphyria are caused by gain of function mutations, while others are characterized by an impaired protein function, causing a metabolic pathway blockage. Pathophysiology classification includes intoxication, storage and energy-related metabolic disorders. Factors specific to each disease trigger acute metabolic decompensations. IEM require prompt and effective care, since therapeutic delay has been associated with the development of fatal events including severe metabolic acidosis, hyperammonemia, cerebral edema, and death. Rapid expression of therapeutic proteins can be achieved hours after the administration of messenger RNAs (mRNA), representing an etiological solution for acute decompensations. mRNA-based therapy relies on modified RNAs with enhanced stability and translatability into therapeutic proteins. The proteins produced in the ribosomes can be targeted to specific intracellular compartments, the cell membrane, or be secreted. Non-immunogenic lipid nanoparticle formulations have been optimized to prevent RNA degradation and to allow safe repetitive administrations depending on the disease physiopathology and clinical status of the patients, thus, mRNA could be also an effective chronic treatment for IEM. Given that the liver plays a key role in most of metabolic pathways or can be used as bioreactor for excretable proteins, this review focuses on the preclinical and clinical evidence that supports the implementation of mRNA technology as a promising personalized strategy for liver metabolic disorders such as acute intermittent porphyria, ornithine transcarbamylase deficiency or glycogen storage disease.


Subject(s)
Liver Diseases , Metabolic Diseases , Metabolism, Inborn Errors , Nanoparticles , Adult , Child , Humans , Liposomes , Metabolic Diseases/complications , Metabolic Diseases/genetics , Metabolic Diseases/therapy , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/genetics , Metabolism, Inborn Errors/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism
4.
J Exp Clin Cancer Res ; 41(1): 183, 2022 May 26.
Article in English | MEDLINE | ID: mdl-35619118

ABSTRACT

BACKGROUND: Cholangiocarcinoma (CCA) is still a deadly tumour. Histological and molecular aspects of thioacetamide (TAA)-induced intrahepatic CCA (iCCA) in rats mimic those of human iCCA. Carcinogenic changes and therapeutic vulnerabilities in CCA may be captured by molecular investigations in bile, where we performed bile proteomic and metabolomic analyses that help discovery yet unknown pathways relevant to human iCCA. METHODS: Cholangiocarcinogenesis was induced in rats (TAA) and mice (JnkΔhepa + CCl4 + DEN model). We performed proteomic and metabolomic analyses in bile from control and CCA-bearing rats. Differential expression was validated in rat and human CCAs. Mechanisms were addressed in human CCA cells, including Huh28-KRASG12D cells. Cell signaling, growth, gene regulation and [U-13C]-D-glucose-serine fluxomics analyses were performed. In vivo studies were performed in the clinically-relevant iCCA mouse model. RESULTS: Pathways related to inflammation, oxidative stress and glucose metabolism were identified by proteomic analysis. Oxidative stress and high amounts of the oncogenesis-supporting amino acids serine and glycine were discovered by metabolomic studies. Most relevant hits were confirmed in rat and human CCAs (TCGA). Activation of interleukin-6 (IL6) and epidermal growth factor receptor (EGFR) pathways, and key genes in cancer-related glucose metabolic reprogramming, were validated in TAA-CCAs. In TAA-CCAs, G9a, an epigenetic pro-tumorigenic writer, was also increased. We show that EGFR signaling and mutant KRASG12D can both activate IL6 production in CCA cells. Furthermore, phosphoglycerate dehydrogenase (PHGDH), the rate-limiting enzyme in serine-glycine pathway, was upregulated in human iCCA correlating with G9a expression. In a G9a activity-dependent manner, KRASG12D promoted PHGDH expression, glucose flow towards serine synthesis, and increased CCA cell viability. KRASG12D CAA cells were more sensitive to PHGDH and G9a inhibition than controls. In mouse iCCA, G9a pharmacological targeting reduced PHGDH expression. CONCLUSIONS: In CCA, we identified new pro-tumorigenic mechanisms: Activation of EGFR signaling or KRAS mutation drives IL6 expression in tumour cells; Glucose metabolism reprogramming in iCCA includes activation of the serine-glycine pathway; Mutant KRAS drives PHGDH expression in a G9a-dependent manner; PHGDH and G9a emerge as therapeutic targets in iCCA.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Animals , Arachnodactyly , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/metabolism , Bile Ducts, Intrahepatic/pathology , Carcinogenesis/genetics , Cholangiocarcinoma/pathology , Contracture , Epigenesis, Genetic , ErbB Receptors/genetics , ErbB Receptors/metabolism , Glucose , Glycine/metabolism , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Mice , Phosphoglycerate Dehydrogenase/genetics , Proteomics , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Rats , Serine/metabolism
5.
Int J Mol Sci ; 22(13)2021 Jun 29.
Article in English | MEDLINE | ID: mdl-34209489

ABSTRACT

The endocytosis of ligand-bound receptors and their eventual recycling to the plasma membrane (PM) are processes that have an influence on signalling activity and therefore on many cell functions, including migration and proliferation. Like other tyrosine kinase receptors (TKR), the insulin receptor (INSR) has been shown to be endocytosed by clathrin-dependent and -independent mechanisms. Once at the early endosome (EE), the sorting of the receptor, either to the late endosome (LE) for degradation or back to the PM through slow or fast recycling pathways, will determine the intensity and duration of insulin effects. Both the endocytic and the endosomic pathways are regulated by many proteins, the Arf and Rab families of small GTPases being some of the most relevant. Here, we argue for a specific role for the slow recycling route, whilst we review the main molecular mechanisms involved in INSR endocytosis, sorting and recycling, as well as their possible role in cell functions.


Subject(s)
Endosomes/metabolism , Receptor, Insulin/metabolism , Animals , Carrier Proteins , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Clathrin/metabolism , Endocytosis , Humans , Lysosomes , Protein Binding , Protein Transport , Receptor, Insulin/agonists , Transport Vesicles/metabolism , rab GTP-Binding Proteins/metabolism
6.
Gut ; 70(2): 388-400, 2021 02.
Article in English | MEDLINE | ID: mdl-32327527

ABSTRACT

OBJECTIVE: Hepatic stellate cells (HSC) transdifferentiation into myofibroblasts is central to fibrogenesis. Epigenetic mechanisms, including histone and DNA methylation, play a key role in this process. Concerted action between histone and DNA-mehyltransferases like G9a and DNMT1 is a common theme in gene expression regulation. We aimed to study the efficacy of CM272, a first-in-class dual and reversible G9a/DNMT1 inhibitor, in halting fibrogenesis. DESIGN: G9a and DNMT1 were analysed in cirrhotic human livers, mouse models of liver fibrosis and cultured mouse HSC. G9a and DNMT1 expression was knocked down or inhibited with CM272 in human HSC (hHSC), and transcriptomic responses to transforming growth factor-ß1 (TGFß1) were examined. Glycolytic metabolism and mitochondrial function were analysed with Seahorse-XF technology. Gene expression regulation was analysed by chromatin immunoprecipitation and methylation-specific PCR. Antifibrogenic activity and safety of CM272 were studied in mouse chronic CCl4 administration and bile duct ligation (BDL), and in human precision-cut liver slices (PCLSs) in a new bioreactor technology. RESULTS: G9a and DNMT1 were detected in stromal cells in areas of active fibrosis in human and mouse livers. G9a and DNMT1 expression was induced during mouse HSC activation, and TGFß1 triggered their chromatin recruitment in hHSC. G9a/DNMT1 knockdown and CM272 inhibited TGFß1 fibrogenic responses in hHSC. TGFß1-mediated profibrogenic metabolic reprogramming was abrogated by CM272, which restored gluconeogenic gene expression and mitochondrial function through on-target epigenetic effects. CM272 inhibited fibrogenesis in mice and PCLSs without toxicity. CONCLUSIONS: Dual G9a/DNMT1 inhibition by compounds like CM272 may be a novel therapeutic strategy for treating liver fibrosis.


Subject(s)
DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Hepatic Stellate Cells/metabolism , Histocompatibility Antigens/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Liver Cirrhosis/etiology , Animals , Chromatin Immunoprecipitation , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Epigenesis, Genetic , Gene Expression Regulation , Gene Knockdown Techniques , Histocompatibility Antigens/genetics , Histone-Lysine N-Methyltransferase/genetics , Humans , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Male , Mice , Mice, Inbred C57BL , Polymerase Chain Reaction , Transforming Growth Factor beta1/metabolism
7.
Cancers (Basel) ; 14(1)2021 Dec 24.
Article in English | MEDLINE | ID: mdl-35008241

ABSTRACT

Fibropolycystic liver disease is characterized by hyperproliferation of the biliary epithelium and the formation of multiple dilated cysts, a process associated with unfolded protein response (UPR). In the present study, we aimed to understand the mechanisms of cyst formation and UPR activation in hepatocytic c-Jun N-terminal kinase 1/2 (Jnk1/2) knockout mice. Floxed JNK1/2 (Jnkf/f) and Jnk∆hepa animals were sacrificed at different time points during progression of liver disease. Histological examination of specimens evidenced the presence of collagen fiber deposition, increased α-smooth muscle actin (αSMA), infiltration of CD45, CD11b and F4/80 cells and proinflammatory cytokines (Tnf, Tgfß1) and liver injury (e.g., ALT, apoptosis and Ki67-positive cells) in Jnk∆hepa compared with Jnkf/f livers from 32 weeks of age. This was associated with activation of effectors of the UPR, including BiP/GRP78, CHOP and spliced XBP1. Tunicamycin (TM) challenge strongly induced ER stress and fibrosis in Jnk∆hepa animals compared with Jnkf/f littermates. Finally, thioacetamide (TAA) administration to Jnk∆hepa mice induced UPR activation, peribiliary fibrosis, liver injury and markers of biliary proliferation and cholangiocarcinoma (CCA). Orthoallografts of DEN/CCl4-treated Jnk∆hepa liver tissue triggered malignant CCA. Altogether, these results suggest that activation of the UPR in conjunction with fibrogenesis might trigger hepatic cystogenesis and early stages of CCA.

8.
Hepatology ; 73(6): 2380-2396, 2021 06.
Article in English | MEDLINE | ID: mdl-33222246

ABSTRACT

BACKGROUND AND AIMS: Cholangiocarcinoma (CCA) is a devastating disease often detected at advanced stages when surgery cannot be performed. Conventional and targeted systemic therapies perform poorly, and therefore effective drugs are urgently needed. Different epigenetic modifications occur in CCA and contribute to malignancy. Targeting epigenetic mechanisms may thus open therapeutic opportunities. However, modifications such as DNA and histone methylation often coexist and cooperate in carcinogenesis. We tested the therapeutic efficacy and mechanism of action of a class of dual G9a histone-methyltransferase and DNA-methyltransferase 1 (DNMT1) inhibitors. APPROACH AND RESULTS: Expression of G9a, DNMT1, and their molecular adaptor, ubiquitin-like with PHD and RING finger domains-1 (UHRF1), was determined in human CCA. We evaluated the effect of individual and combined pharmacological inhibition of G9a and DNMT1 on CCA cell growth. Our lead G9a/DNMT1 inhibitor, CM272, was tested in human CCA cells, patient-derived tumoroids and xenograft, and a mouse model of cholangiocarcinogenesis with hepatocellular deletion of c-Jun-N-terminal-kinase (Jnk)-1/2 and diethyl-nitrosamine (DEN) plus CCl4 treatment (JnkΔhepa + DEN + CCl4 mice). We found an increased and correlative expression of G9a, DNMT1, and UHRF1 in CCAs. Cotreatment with independent pharmacological inhibitors G9a and DNMT1 synergistically inhibited CCA cell growth. CM272 markedly reduced CCA cell proliferation and synergized with Cisplatin and the ERBB-targeted inhibitor, Lapatinib. CM272 inhibited CCA tumoroids and xenograft growth and significantly antagonized CCA progression in JnkΔhepa + DEN + CCl4 mice without apparent toxicity. Mechanistically, CM272 reprogrammed the tumoral metabolic transcriptome and phenotype toward a differentiated and quiescent status. CONCLUSIONS: Dual targeting of G9a and DNMT1 with epigenetic small molecule inhibitors such as CM272 is a potential strategy to treat CCA and/or enhance the efficacy of other systemic therapies.


Subject(s)
Bile Duct Neoplasms , Cell Proliferation/drug effects , Cholangiocarcinoma , DNA (Cytosine-5-)-Methyltransferase 1 , Enzyme Inhibitors/pharmacology , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase , Animals , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/metabolism , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation/drug effects , DNA Methylation/physiology , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Histocompatibility Antigens/metabolism , Histone Code/drug effects , Histone Code/physiology , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/metabolism , Humans , Mice , Treatment Outcome , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays/methods
9.
Cancers (Basel) ; 12(6)2020 Jun 21.
Article in English | MEDLINE | ID: mdl-32575903

ABSTRACT

Cholangiocarcinoma (CCA) and pancreatic adenocarcinoma (PDAC) may lead to the development of extrahepatic obstructive cholestasis. However, biliary stenoses can also be caused by benign conditions, and the identification of their etiology still remains a clinical challenge. We performed metabolomic and proteomic analyses of bile from patients with benign (n = 36) and malignant conditions, CCA (n = 36) or PDAC (n = 57), undergoing endoscopic retrograde cholangiopancreatography with the aim of characterizing bile composition in biliopancreatic disease and identifying biomarkers for the differential diagnosis of biliary strictures. Comprehensive analyses of lipids, bile acids and small molecules were carried out using mass spectrometry (MS) and nuclear magnetic resonance spectroscopy (1H-NMR) in all patients. MS analysis of bile proteome was performed in five patients per group. We implemented artificial intelligence tools for the selection of biomarkers and algorithms with predictive capacity. Our machine-learning pipeline included the generation of synthetic data with properties of real data, the selection of potential biomarkers (metabolites or proteins) and their analysis with neural networks (NN). Selected biomarkers were then validated with real data. We identified panels of lipids (n = 10) and proteins (n = 5) that when analyzed with NN algorithms discriminated between patients with and without cancer with an unprecedented accuracy.

10.
Int J Mol Sci ; 21(4)2020 Feb 19.
Article in English | MEDLINE | ID: mdl-32092977

ABSTRACT

AGAP2 (Arf GAP with GTP-binding protein-like domain, Ankyrin repeat and PH domain 2) isoform 2 is a protein that belongs to the Arf GAP (GTPase activating protein) protein family. These proteins act as GTPase switches for Arfs, which are Ras superfamily members, being therefore involved in signaling regulation. Arf GAP proteins have been shown to participate in several cellular functions including membrane trafficking and actin cytoskeleton remodeling. AGAP2 is a multi-tasking Arf GAP that also presents GTPase activity and is involved in several signaling pathways related with apoptosis, cell survival, migration, and receptor trafficking. The increase of AGAP2 levels is associated with pathologies as cancer and fibrosis. Transforming growth factor beta-1 (TGF-ß1) is the most potent pro-fibrotic cytokine identified to date, currently accepted as the principal mediator of the fibrotic response in liver, lung, and kidney. Recent literature has described that the expression of AGAP2 modulates some of the pro-fibrotic effects described for TGF-ß1 in the liver. The present review is focused on the interrelated molecular effects between AGAP2 and TGFß1 expression, presenting AGAP2 as a new player in the signaling of this pro-fibrotic cytokine, thereby contributing to the progression of hepatic fibrosis.


Subject(s)
GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/metabolism , Hepatic Stellate Cells/metabolism , Liver Cirrhosis/metabolism , Signal Transduction/genetics , Transforming Growth Factor beta1/metabolism , Cell Differentiation/genetics , Cell Movement/genetics , Cell Proliferation/genetics , GTP-Binding Proteins/genetics , GTPase-Activating Proteins/genetics , Hepatic Stellate Cells/enzymology , Humans , Liver Cirrhosis/enzymology , Liver Cirrhosis/genetics , Protein Isoforms/metabolism , Protein Transport/genetics , Transforming Growth Factor beta1/genetics
11.
Biochim Biophys Acta Mol Cell Res ; 1866(4): 673-685, 2019 04.
Article in English | MEDLINE | ID: mdl-30660615

ABSTRACT

Liver damage induces hepatic stellate cells (HSC) activation, characterised by a fibrogenic, proliferative and migratory phenotype. Activated HSC are mainly regulated by transforming growth factor ß 1 (TGFß1), which increases the production of extracellular matrix proteins (e.g. collagen-I) promoting the progression of hepatic fibrosis. AGAP2 (ArfGAP with GTPase domain, ankyrin repeat and PH domain 2) is a GTPase/GTP-activating protein involved in the actin remodelling system and receptor recycling. In the present work the role of AGAP2 in human HSC in response to TGFß1 was investigated. LX-2 HSC were transfected with AGAP2 siRNA and treated with TGFß1. AGAP2 knockdown prevented to some extent the proliferative and migratory TGFß1-induced capacities of LX-2 cells. An array focused on human fibrosis revealed that AGAP2 knockdown partially prevented TGFß1-mediated gene expression of the fibrogenic genes ACTA2, COL1A2, EDN1, INHBE, LOX, PDGFB, TGFΒ12, while favored the expression of CXCR4, IL1A, MMP1, MMP3 and MMP9 genes. Furthermore, TGFß1 induced AGAP2 promoter activation and its protein expression in LX-2. Moreover, AGAP2 protein levels were significantly increased in liver samples from rats with thioacetamide-induced fibrosis. In addition, AGAP2 silencing affected TGFß1-receptor 2 (TGFR2) trafficking in U2OS cells, blocking its effective recycling to the membrane. AGAP2 silencing in LX-2 cells prevented the TGFß1-induced increase of collagen-I protein levels, while its overexpression enhanced collagen-I protein expression in the presence or absence of the cytokine. AGAP2 overexpression also increased focal adhesion kinase (FAK) phosphorylated levels in LX-2 cells. FAK and MEK1 inhibitors prevented the increase of collagen-I expression caused by TGFß1 in LX-2 overexpressing AGAP2. In summary, the present work shows for the first time, that AGAP2 is a potential new target involved in TGFß1 signalling, contributing to the progression of hepatic fibrosis.


Subject(s)
GTP-Binding Proteins/physiology , GTPase-Activating Proteins/physiology , Hepatic Stellate Cells/metabolism , Transforming Growth Factor beta1/physiology , Animals , Cell Line , Cell Movement , Cell Proliferation , Cell Survival , Collagen Type I/metabolism , Focal Adhesion Protein-Tyrosine Kinases/physiology , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/enzymology , Hepatic Stellate Cells/physiology , Humans , Liver Cirrhosis/metabolism , Male , Rats, Sprague-Dawley , Receptor, Transforming Growth Factor-beta Type II/metabolism
12.
Hepatology ; 69(2): 587-603, 2019 02.
Article in English | MEDLINE | ID: mdl-30014490

ABSTRACT

Epigenetic modifications such as DNA and histone methylation functionally cooperate in fostering tumor growth, including that of hepatocellular carcinoma (HCC). Pharmacological targeting of these mechanisms may open new therapeutic avenues. We aimed to determine the therapeutic efficacy and potential mechanism of action of our dual G9a histone-methyltransferase and DNA-methyltransferase 1 (DNMT1) inhibitor in human HCC cells and their crosstalk with fibrogenic cells. The expression of G9a and DNMT1, along with that of their molecular adaptor ubiquitin-like with PHD and RING finger domains-1 (UHRF1), was measured in human HCCs (n = 268), peritumoral tissues (n = 154), and HCC cell lines (n = 32). We evaluated the effect of individual and combined inhibition of G9a and DNMT1 on HCC cell growth by pharmacological and genetic approaches. The activity of our lead compound, CM-272, was examined in HCC cells under normoxia and hypoxia, human hepatic stellate cells and LX2 cells, and xenograft tumors formed by HCC or combined HCC+LX2 cells. We found a significant and correlative overexpression of G9a, DNMT1, and UHRF1 in HCCs in association with poor prognosis. Independent G9a and DNMT1 pharmacological targeting synergistically inhibited HCC cell growth. CM-272 potently reduced HCC and LX2 cells proliferation and quelled tumor growth, particularly in HCC+LX2 xenografts. Mechanistically, CM-272 inhibited the metabolic adaptation of HCC cells to hypoxia and induced a differentiated phenotype in HCC and fibrogenic cells. The expression of the metabolic tumor suppressor gene fructose-1,6-bisphosphatase (FBP1), epigenetically repressed in HCC, was restored by CM-272. Conclusion: Combined targeting of G9a/DNMT1 with compounds such as CM-272 is a promising strategy for HCC treatment. Our findings also underscore the potential of differentiation therapy in HCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , Histone-Lysine N-Methyltransferase/antagonists & inhibitors , Liver Neoplasms, Experimental/drug therapy , Animals , Antineoplastic Agents/pharmacology , CCAAT-Enhancer-Binding Proteins/metabolism , Carcinoma, Hepatocellular/enzymology , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Dogs , Hep G2 Cells , Histone-Lysine N-Methyltransferase/metabolism , Humans , Liver Neoplasms, Experimental/enzymology , Madin Darby Canine Kidney Cells , Male , Mice, Nude , Ubiquitin-Protein Ligases/metabolism , Xenograft Model Antitumor Assays
13.
Free Radic Biol Med ; 126: 15-26, 2018 10.
Article in English | MEDLINE | ID: mdl-30036633

ABSTRACT

NADPH oxidase (Nox) variants Nox1, Nox2 and Nox4 are implicated in the progression of liver fibrosis. However, the role of Nox5 is not yet known, mainly due to the lack of this enzyme in rat and mouse genomes. Here we describe the expression and functional relevance of Nox5 in the human cell line of hepatic stellate cells (HSC) LX-2. Under basal conditions, three long (Nox5-L: Nox5α, -ß, and -δ) and a short (Nox5-S or Nox5ε) splice variants were detected, which were silenced with specific siRNAs for Nox5. The most abundant isoform was Nox5-S, accounting for more than 90% of Nox5 protein. Overexpression of Nox5ß generated reactive oxygen species (ROS) in the presence of calcium, as judged by the production of hydrogen peroxide, L-012 luminescence and cytochrome c reduction. Nox5ε did not generated ROS under these conditions, and a reduced ROS production was observed when co-expressed with Nox5ß. In contrast, dihydroethidium oxidation was increased by Nox5ß or Nox5ε, suggesting that Nox5ε induced intracellular oxidative stress by an unknown mechanism. Functional studies showed that both Nox5ß and Nox5ε stimulated the proliferation of LX-2 cells and the collagen type I levels, while Nox5 siRNAs inhibited these effects. Interestingly, TGF-ß and angiotensin II upregulated Nox5 expression, which was reduced in cells pre-incubated with catalase. Further studies silencing Nox5 in TGF-ß-treated cells resulted in a reduction of collagen levels via p38 MAPK. Collectively, these results show for the first time that Nox5 can play a relevant role in the proliferation and fibrosis on human HSC.


Subject(s)
Hepatic Stellate Cells/enzymology , Liver Cirrhosis/enzymology , NADPH Oxidase 5/genetics , Protein Isoforms/genetics , Cell Line , Cell Proliferation/genetics , Gene Expression Regulation, Enzymologic , Humans , Liver Cirrhosis/physiopathology , NADPH Oxidase 5/metabolism , Oxidation-Reduction , Oxidative Stress/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
14.
Biochim Biophys Acta ; 1863(8): 2115-23, 2016 08.
Article in English | MEDLINE | ID: mdl-27155082

ABSTRACT

Unfolded protein response (UPR) triggered as a consequence of ER stress has been shown to be involved in the development of different pathologies, including fibrotic disorders. In the present paper we explore the role played by UPR on a key fibrogenic parameter in the liver: collagen type I levels in activated hepatic stellate cells (HSC). Using Brefeldin A (BFA) as an ER stress inducer we found that UPR correlated with enhanced mRNA and protein levels of collagen type I in a cell line of immortalized non-tumoral rat HSC. Analysis of the three branches of UPR revealed the activation of IRE1α, PERK and ATF6 in response to BFA, although PERK activation was shown not to be involved in the fibrogenic action of BFA. BFA also activated p38 MAPK in an IRE1α-dependent way and the p38 MAPK inhibitor SB203580 prevented the increase in collagen type I mRNA and protein levels caused by BFA, suggesting the involvement of this kinase on this effect. Analysis of Smad activation showed that phosphorylated nuclear levels of Smad2 and 3 were increased in response to BFA treatment. Inhibition of Smad3 phosphorylation by SIS3 prevented the enhancement of collagen type I levels caused by BFA. Pretreatment with IRE1α and p38 MAPK inhibitors also prevented the increased p-Smad3 accumulation in the nucleus, suggesting an IRE1α-p38 MAPK-Smad pathway to be responsible for the fibrogenic action of BFA on HSC.


Subject(s)
Brefeldin A/pharmacology , Collagen Type I/biosynthesis , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/physiology , Hepatic Stellate Cells/drug effects , MAP Kinase Signaling System/drug effects , Multienzyme Complexes/physiology , Protein Serine-Threonine Kinases/physiology , Smad3 Protein/physiology , Unfolded Protein Response/drug effects , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Cell Line , Collagen Type I/genetics , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/antagonists & inhibitors , Fibrosis , Gene Expression Regulation/drug effects , Hepatic Stellate Cells/metabolism , Imidazoles/pharmacology , Multienzyme Complexes/antagonists & inhibitors , Phosphorylation , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyridines/pharmacology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Unfolded Protein Response/physiology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
15.
Free Radic Biol Med ; 87: 169-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26119779

ABSTRACT

Apigenin, a natural flavone, is emerging as a promising compound for the treatment of several diseases. One of the hallmarks of apigenin is the generation of intracellular reactive oxygen species (ROS), as judged by the oxidation of reduced dichlorofluorescein derivatives seen in many cell types. This study aimed to reveal some mechanisms by which apigenin can be oxidized and how apigenin-derived radicals affect the oxidation of 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein (H(2)DCF), a probe usually employed to detect intracellular ROS. Apigenin induced a rapid oxidation of H(2)DCF in two different immortalized cell lines derived from rat and human hepatic stellate cells. However, apigenin did not generate ROS in these cells, as judged by dihydroethidium oxidation and extracellular hydrogen peroxide production. In cell-free experiments we found that oxidation of apigenin leads to the generation of a phenoxyl radical, which directly oxidizes H(2)DCF with catalytic amounts of hydrogen peroxide. The net balance of the reaction was the oxidation of the probe by molecular oxygen due to redox cycling of apigenin. This flavonoid was also able to deplete NADH and glutathione by a similar mechanism. Interestingly, H(2)DCF oxidation was significantly accelerated by apigenin in the presence of horseradish peroxidase and xanthine oxidase, but not with other enzymes showing peroxidase-like activity, such as cytochrome c or catalase. We conclude that in cells treated with apigenin oxidation of reduced dichlorofluorescein derivatives does not measure intracellular ROS and that pro- and antioxidant effects of flavonoids deduced from these experiments are inconclusive and must be confirmed by other techniques.


Subject(s)
Antioxidants/administration & dosage , Apigenin/administration & dosage , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Animals , Cell Line , Free Radicals/metabolism , Glutathione/metabolism , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Humans , Hydrogen Peroxide/metabolism , Oxidation-Reduction/drug effects , Oxygen/metabolism , Rats
16.
J Cell Physiol ; 230(3): 546-53, 2015 Mar.
Article in English | MEDLINE | ID: mdl-24976518

ABSTRACT

The turnover of extracellular matrix (ECM) components can generate signals that regulate several cellular functions such as proliferation, differentiation, and apoptosis. During liver injury, matrix metalloproteases (MMPs) production is enhanced and increased levels of peptides derived from extracellular matrix proteins can be generated. Synthetic peptides with sequences present in extracellular matrix proteins were previously found to induce both stimulating and apoptotic effects on several cell types including the inflammatory cells monocytes/macrophages. Therefore, in inflammatory liver diseases, locally accumulated peptides could be also important in regulating hepatic fibrosis by inducing apoptosis of hepatic stellate cells (HSC), the primary cellular source of extracellular matrix components. Here, we describe the apoptotic effect of fibronectin peptides on the cell line of human hepatic stellate cells LX-2 based on oligonucleosomal DNA fragmentation, caspase-3 and -9 activation, Bcl-2 depletion, and accumulation of Bax protein. We also found that these peptides trigger the activation of Src kinase, which in turn mediated the increase of JNK and p38 activities. By the use of specific inhibitors we demonstrated the involvement of Src, JNK, and p38 in apoptosis induced by fibronectin peptides on HSC. Moreover, fibronectin peptides increased iNOS expression in human HSC, and specific inhibition of iNOS significantly reduced the sustained activity of JNK and the programmed cell death caused by these peptides. Finally, the possible regulatory effect of fibronectin peptides in liver fibrosis was further supported by the ability of these peptides to induce metalloprotease-9 (MMP-9) expression in human monocytes.


Subject(s)
Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Liver Cirrhosis/metabolism , Peptides/metabolism , Apoptosis/genetics , Caspase 3/metabolism , Cell Differentiation/genetics , Cell Line , Cell Proliferation/genetics , DNA Fragmentation , Fibronectins/genetics , Gene Expression Regulation , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Liver Cirrhosis/pathology , Matrix Metalloproteinase 9/metabolism , bcl-2-Associated X Protein/metabolism
17.
Lab Invest ; 93(3): 303-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23318883

ABSTRACT

General control nonderepresible 2 (GCN2) is a highly conserved cytosolic kinase that modulates a complex response for coping with the stress owing to lack of amino acids. GCN2 has been recently shown to be involved in the regulation of metabolic balance and lipid degradation rate in the liver. We hypothesized that GCN2 could have a role in in hepatic fibrogenesis and in the response to acute or chronic liver injury. Activation of GCN2 in primary or immortalized human hepatic stellate cells by incubation with medium lacking the essential amino acid histidine correlated with decreased levels of collagen type I protein and mRNA, suggesting an antifibrogenic effect of GCN2. In vivo studies with Gcn2 knock-out mice (Gcn2(-/-)) showed increased susceptibility to both acute or chronic liver damage induced by CCl(4), as shown by higher alanine aminotransferase and aspartate aminotransferase activities, increased necrosis and higher inflammatory infiltrates compared with wild-type mice (WT). Chronic CCl(4) treatment increased deposition of interstitial collagen type I more in Gcn2(-/-) mice than in WT mice. Col1a1 and col1a2 mRNA levels also increased in CCl(4)-treated Gcn2(-/-) mice compared with WT mice. These results suggest that GCN2 is a key regulator of the fibrogenic response to liver injury.


Subject(s)
Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/enzymology , Liver Cirrhosis/enzymology , Protein Serine-Threonine Kinases/metabolism , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Blotting, Western , Chemical and Drug Induced Liver Injury/complications , Chemical and Drug Induced Liver Injury/pathology , Collagen Type I/metabolism , Culture Media/chemistry , DNA Primers/genetics , Enzyme Activation/physiology , Hepatic Stellate Cells/enzymology , Histidine/deficiency , Humans , Immunohistochemistry , Liver Cirrhosis/etiology , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Protein Serine-Threonine Kinases/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
18.
Biochem Pharmacol ; 81(3): 451-8, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21056031

ABSTRACT

Inflammatory conditions are characterized by continuous overproduction of nitric oxide (NO) that can contribute to cell survival but also to cell demise by affecting apoptosis. These facts are important in regulation of hepatic fibrogenesis during exposure to inflammatory stress, since elevated NO may pose the risk of cells with a pro-fibrogenic phenotype giving rise to a sustained proliferation leading to chronic fibrosis. Since nitration of tyrosine residues occurs in a range of diseases involving inflammation, we tested the hypothesis that nitration of specific proteins could result in apoptosis of hepatic stellate cells (HSC), the primary cellular source of matrix components in liver diseases. We found the peroxynitrite generator SIN-1 to promote apoptosis in human and rat HSC, based on oligonucleosomal DNA fragmentation, caspase-3 and -9 activation, Bcl-2 depletion and accumulation of Bax protein. We also showed that SIN-1-induced apoptosis of HSC was due to protein nitration. Among the tyrosine-nitrated proteins, tyrosine kinase Lyn was identified. SIN-1 triggered a signaling pathway through Src kinase Lyn activation that resulted in increased activity of the tyrosine kinase Syk. The involvement of these signaling molecules in the apoptotic process induced by SIN-1 as well as the mechanism by which they are activated was confirmed by using specific inhibitors. In summary, NO, via protein-nitration, could play an important role in controlling liver fibrosis resolution by regulation of HSC apoptosis.


Subject(s)
Apoptosis , Hepatic Stellate Cells/metabolism , Molsidomine/analogs & derivatives , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Animals , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line , DNA Fragmentation , Humans , Inflammation/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Liver Cirrhosis/metabolism , Molsidomine/metabolism , Molsidomine/pharmacology , Protein-Tyrosine Kinases/metabolism , Rats , Signal Transduction , Syk Kinase , Tyrosine/metabolism , src-Family Kinases/metabolism
19.
Cell Physiol Biochem ; 26(3): 281-90, 2010.
Article in English | MEDLINE | ID: mdl-20798512

ABSTRACT

In eukaryotic cells amino acid deprivation triggers a response aimed to ensure cell survival in stress conditions. In the present work we analyzed the effects of amino acid deprivation on intracellular levels of reactive oxygen species (ROS) of hepatic stellate cells (HSC), a key cell type in the development of liver fibrosis. Histidine deprivation caused in the human immortalized HSC cell line LX-2 a fast decrease of intracellular ROS levels that was also observed in HSC incubated either with leucine-free or amino acid-free medium, but not with glucose-free medium. Phosphorylation of GCN2 kinase and its substrate eIF2alpha was induced by histidine deprivation. Reversion studies and activation of GCN2 by tRNA and the proteasome inhibitor MG-132 showed a correlation between GCN2 phosphorylation and diminished ROS levels. However, a lack of correlation between eIF2alpha phosphorylation and ROS levels was found using salubrinal, an inhibitor of eIF2alpha phosphorylation, suggesting a role for GCN2 unrelated to its activity as eIF2alpha kinase. LX-2 cells treated with histidine-free medium presented reduced SOD activity that could account for the decrease on ROS levels. Histidine deprivation as well as activation of GCN2 by treatment with tRNA, caused an increase in LX-2 cell viability, suggesting amino acid restriction to present a protective effect in HSC which is mediated by GCN2 activation.


Subject(s)
Amino Acids/physiology , Hepatic Stellate Cells/metabolism , Reactive Oxygen Species/metabolism , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Humans , Leupeptins/pharmacology , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Superoxide Dismutase/metabolism
20.
Apoptosis ; 13(11): 1356-67, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18819005

ABSTRACT

The sustained overproduction of nitric oxide (NO) observed in inflammatory conditions can contribute to cell demise by affecting apoptosis. Nitration of tyrosine residues occurs in a range of diseases involving macrophage activation. Since NO induces apoptosis in monocytes/macrophages, we tested the hypothesis that nitration of specific proteins could result in apoptotic cell death. The peroxynitrite generator SIN-1 promoted apoptosis in monocytes based on oligonucleosomal DNA fragmentation, caspase-3 and -9 activation, Bcl-2 depletion and accumulation of Bax and p53 proteins. We also found that the signaling pathway triggered by SIN-1 was initiated through tyrosine kinase and Rac activation and resulted in increased JNK and p38 activities. Among the tyrosine-nitrated proteins, Rac and Lyn were identified. Using specific inhibitors for different signaling and effector molecules involved in the apoptotic process we demonstrate that NO, via protein-nitration, could play an important role in controlling the inflammatory response by regulation of monocyte homeostasis.


Subject(s)
Apoptosis , Monocytes/metabolism , Nitric Oxide/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , DNA Fragmentation , Humans , Inflammation , Leukocytes, Mononuclear/metabolism , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System , Macrophages/metabolism , Models, Biological , Nitrogen/chemistry , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...