Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
BMC Cancer ; 16: 9, 2016 Jan 11.
Article in English | MEDLINE | ID: mdl-26753564

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality. We previously showed that the inhibition of placental growth factor (PlGF) exerts antitumour effects and induces vessel normalisation, possibly reducing hypoxia. However, the exact mechanism underlying these effects remains unclear. Because hypoxia and endoplasmic reticulum stress, which activates the unfolded protein response (UPR), have been implicated in HCC progression, we assessed the interactions between PlGF and these microenvironmental stresses. METHODS: PlGF knockout mice and validated monoclonal anti-PlGF antibodies were used in a diethylnitrosamine-induced mouse model for HCC. We examined the interactions among hypoxia, UPR activation and PlGF induction in HCC cells. RESULTS: Both the genetic and pharmacological inhibitions of PlGF reduced the chaperone levels and the activation of the PKR-like endoplasmic reticulum kinase (PERK) pathway of the UPR in diethylnitrosamine-induced HCC. Furthermore, we identified that tumour hypoxia was attenuated, as shown by reduced pimonidazole binding. Interestingly, hypoxic exposure markedly activated the PERK pathway in HCC cells in vitro, suggesting that PlGF inhibition may diminish PERK activation by improving oxygen delivery. We also found that PlGF expression is upregulated by different chemical UPR inducers via activation of the inositol-requiring enzyme 1 pathway in HCC cells. CONCLUSIONS: PlGF inhibition attenuates PERK activation, likely by tempering hypoxia in HCC via vessel normalisation. The UPR, in turn, is able to regulate PlGF expression, suggesting the existence of a feedback mechanism for hypoxia-mediated UPR that promotes the expression of the angiogenic factor PlGF. These findings have important implications for our understanding of the effect of therapies normalising tumour vasculature.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Neovascularization, Pathologic/genetics , Pregnancy Proteins/biosynthesis , eIF-2 Kinase/biosynthesis , Animals , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/pathology , Cell Hypoxia/drug effects , Cell Hypoxia/genetics , Diethylnitrosamine/toxicity , Disease Models, Animal , Endoplasmic Reticulum Stress/genetics , Gene Expression Regulation, Neoplastic/drug effects , Glycosaminoglycans/physiology , Hep G2 Cells , Humans , Liver Neoplasms/chemically induced , Liver Neoplasms/pathology , Mice , Mice, Knockout , Neovascularization, Pathologic/pathology , Placenta Growth Factor , Pregnancy Proteins/genetics , Tumor Microenvironment/genetics , Unfolded Protein Response/genetics , eIF-2 Kinase/genetics
2.
Oncotarget ; 6(29): 28011-25, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26293671

ABSTRACT

Hepatocellular carcinoma (HCC) is characterized by the accumulation of unfolded proteins in the endoplasmic reticulum (ER), which activates the unfolded protein response (UPR). However, the role of ER stress in tumor initiation and progression is controversial. To determine the impact of ER stress, we applied tauroursodeoxycholic acid (TUDCA), a bile acid with chaperone properties. The effects of TUDCA were assessed using a diethylnitrosamine-induced mouse HCC model in preventive and therapeutic settings. Cell metabolic activity, proliferation and invasion were investigated in vitro. Tumor progression was assessed in the HepG2 xenograft model. Administration of TUDCA in the preventive setting reduced carcinogen-induced elevation of alanine and aspartate aminotransferase levels, apoptosis of hepatocytes and tumor burden. TUDCA also reduced eukaryotic initiation factor 2α (eIf2α) phosphorylation, C/EBP homologous protein expression and caspase-12 processing. Thus, TUDCA suppresses carcinogen-induced pro-apoptotic UPR. TUDCA alleviated hepatic inflammation by increasing NF-κB inhibitor IκBα. Furthermore, TUDCA altered the invasive phenotype and enhanced metabolic activity but not proliferation in HCC cells. TUDCA administration after tumor development did not alter orthotopic tumor or xenograft growth. Taken together, TUDCA attenuates hepatocarcinogenesis by suppressing carcinogen-induced ER stress-mediated cell death and inflammation without stimulating tumor progression. Therefore, this chemical chaperone could represent a novel chemopreventive agent.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/pathology , Taurochenodeoxycholic Acid/pharmacology , Animals , Blotting, Western , Carcinogens/toxicity , Disease Models, Animal , Hep G2 Cells , Humans , Immunohistochemistry , In Situ Nick-End Labeling , Male , Mice , Real-Time Polymerase Chain Reaction , Xenograft Model Antitumor Assays
3.
PLoS One ; 10(3): e0119555, 2015.
Article in English | MEDLINE | ID: mdl-25793288

ABSTRACT

BACKGROUND & AIMS: Expression of liver progenitor cell (LPC) characteristics has been proposed as a negative prognostic marker in primary liver tumors. Hypoxia has been linked to activation of the Notch pathway which is responsible for activation and proliferation of LPCs and hypoxia-induced LPC activation has been shown in hepatocellular carcinoma. Our aim was to elucidate the time-dependent effects of hypoxia on the LPC niche in hepatocellular carcinoma which could aid in determining a safe time frame for use of hypoxia inducing therapies. METHODS: We used dimethyloxaloylglycine to mimic a hypoxic reaction in mice by stabilizing hypoxia-inducible factor 1 alpha at three distinct time points in diethylnitrosamine induced hepatocarcinogenesis. LPC, metastasis and Notch pathway markers were determined by quantitative PCR and (immune)histochemistry (heamatoxillin-eosin, reticulin, Sirius red and cytokeratin 19 staining). RESULTS: Activating the hypoxia inducible pathway early in hepatocarcinogenesis resulted in an increased incidence of both cholangioma and hepatocellular lesions, associated with high expression of LPC, metastatic and Notch pathway markers. Adversely, activating the hypoxic response during tumor development resulted in decreased incidence of hepatocellular lesions and increased cholangioma incidence, with an unaltered gene expression profile of LPC-, Notch pathway- and metastatic markers. A hypoxic insult at advanced stages of hepatocarcinogenesis severely increased the expression of LPC characteristics, however without increased expression of actors of the Notch pathway and metastatic markers and minor changes in incidence of hepatocellular and cholangioma lesions. CONCLUSION: Our results indicate that increased hypoxia at the onset of tumor development has detrimental effects on tumor progression; patients with HCC developed in a background of fibrosis/cirrhosis might therefore represent a more difficult treatment group. In contrast, hypoxia during tumor development appears to favor tumor outcome, highlighting the importance of early detection. Finally, hypoxia in advanced stages resulted in increased expression of LPC characteristics indicating poor outcome.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Hypoxia/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Stem Cells/metabolism , Animals , Antigens, Neoplasm/metabolism , Biomarkers , Carcinoma, Hepatocellular/genetics , Cell Adhesion Molecules/metabolism , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Epithelial Cell Adhesion Molecule , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunohistochemistry , Keratin-19/metabolism , Liver Neoplasms/genetics , Male , Mice , Neoplasm Metastasis , RNA, Messenger/genetics , Receptors, Notch/metabolism , Time Factors
4.
Hepatol Int ; 9(1): 93-104, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25598862

ABSTRACT

BACKGROUND: Functional disturbances of the endoplasmic reticulum (ER) lead to activation of the unfolded protein response (UPR), which is involved in the consecutive steps of carcinogenesis. In human hepatocellular carcinoma (HCC), the UPR is shown to be activated; however, little is known about the UPR kinetics and effects of UPR modulation in HCC. METHODS: We sequentially monitored the UPR over time in an orthotopic mouse model for HCC and explored the effects of UPR modulation on cell viability and proliferation in vitro and in the mouse model. RESULTS: The expression of ER-resident chaperones peaked during tumor initiation and increased further during tumor progression, predominantly within the nodules. A peak in Ire1 signaling was observed during tumor initiation. The Perk pathway was activated during tumor progression, and the proapoptotic target Chop was upregulated from week 5 and continued to rise, especially in the tumors. The Atf6 pathway was modestly activated only after tumor initiation. Consistent with the UPR activation, electron microscopy demonstrated ER expansion and reorganization in HCC cells in vivo. Strikingly, under ER stress or hypoxia, the Perk inhibitor and not the Ire1 inhibitor reduced cell viability and proliferation via escalating proteotoxic stress in vitro. Notably, the Perk inhibitor significantly decreased tumor burden in the mouse model. CONCLUSION: We provide the first evaluation of the UPR dynamics in a long-term cancer model and identified a small molecule inhibitor of Perk as a promising strategy for HCC therapy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Transformation, Neoplastic/metabolism , Liver Neoplasms, Experimental/metabolism , Protein Kinase Inhibitors/pharmacology , Unfolded Protein Response , eIF-2 Kinase/metabolism , Activating Transcription Factor 6/genetics , Adaptation, Physiological/drug effects , Animals , Carcinoma, Hepatocellular/chemistry , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/ultrastructure , Cell Proliferation/drug effects , Cell Survival/drug effects , Endoplasmic Reticulum , Endoplasmic Reticulum Chaperone BiP , HSP40 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Hep G2 Cells , Humans , Liver Neoplasms, Experimental/chemistry , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/ultrastructure , Male , Membrane Glycoproteins/genetics , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Mice , Oxidative Stress , Phosphorylation/drug effects , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA, Messenger/analysis , Signal Transduction , Transcription Factor CHOP/analysis , Transcription Factor CHOP/genetics , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , eIF-2 Kinase/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...