Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Pak J Biol Sci ; 25(1): 15-22, 2022 Jan.
Article in English | MEDLINE | ID: mdl-35001571

ABSTRACT

<b>Background and Objective:</b> Pineapple (<i>Ananas comosus</i>) is a popular fruit worldwide with natural antioxidant properties. This study examined how pineapple modified the expression of drug-metabolizing enzymes (CYP1A2, CYP2C9, CYP3A4, UGT1A6, NAT2 and SULT1A1) and a drug transporter (OATP1B1) in human hepatocarcinoma (HepG2) cells. <b>Materials and Methods:</b> HepG2 cells (2.5×10<sup>5</sup> cells/well in a 24-well plate) were incubated with pineapple juice extract (125-1,000 µg mL<sup>1</sup>) for 48 hrs in phenol red-free medium. Resazurin reduction, ROS, AST and ALT assays were performed. The mRNA expression of target genes was determined by RT/qPCR. <b>Results:</b> Pineapple juice slightly reduced HepG2 cell viability to 80% of the control, while ROS, AST and ALT levels were not changed. Pineapple juice did not alter the expression of CYP1A2, CYP2C9 and UGT1A6 mRNA. All tested concentrations of pineapple juice suppressed CYP3A4, NAT2 and OATP1B1 expression, while SULT1A1 expression was induced. <b>Conclusion:</b> Though pineapple juice slightly decreased the viability of HepG2 cells, cell morphology and cell function remained normal. Pineapple juice disturbed the expression of phase I (CYP3A4) and phase II (NAT2 and SULT1A1) metabolizing genes and the drug transporter OATP1B1. Therefore, the consumption of excessive amounts of pineapple juice poses a risk for drug interactions.


Subject(s)
Ananas/metabolism , Fruit and Vegetable Juices/standards , Gene Expression/drug effects , Hep G2 Cells/drug effects , Ananas/microbiology , Arylamine N-Acetyltransferase/drug effects , Arylamine N-Acetyltransferase/genetics , Arylsulfotransferase/drug effects , Arylsulfotransferase/genetics , Cytochrome P-450 CYP3A/drug effects , Cytochrome P-450 CYP3A/genetics , Hep G2 Cells/physiology , Humans
2.
Hepatology ; 73(4): 1307-1326, 2021 04.
Article in English | MEDLINE | ID: mdl-32557804

ABSTRACT

BACKGROUND AND AIMS: Fat accumulation results from increased fat absorption and/or defective fat metabolism. Currently, the lipid-sensing nuclear receptor that controls fat utilization in hepatocytes is elusive. Liver X receptor alpha (LXRα) promotes accumulation of lipids through the induction of several lipogenic genes. However, its effect on lipid degradation is open for study. Here, we investigated the inhibitory role of LXRα in autophagy/lipophagy in hepatocytes and the underlying basis. APPROACH AND RESULTS: In LXRα knockout mice fed a high-fat diet, or cell models, LXRα activation suppressed the function of mitochondria by inhibiting autophagy/lipophagy and induced hepatic steatosis. Gene sets associated with "autophagy" were enriched in hepatic transcriptome data. Autophagy flux was markedly augmented in the LXRα knockout mouse liver and primary hepatocytes. Mechanistically, LXRα suppressed autophagy-related 4B cysteine peptidase (ATG4B) and Rab-8B, responsible for autophagosome and -lysosome formation, by inducing let-7a and microRNA (miR)-34a. Chromatin immunoprecipitation assay enabled us to find LXRα as a transcription factor of let-7a and miR-34a. Moreover, 3' untranslated region luciferase assay substantiated the direct inhibitory effects of let-7a and miR-34a on ATG4B and Rab-8B. Consistently, either LXRα activation or the let-7a/miR-34a transfection lowered mitochondrial oxygen consumption rate and mitochondrial transmembrane potential and increased fat levels. In obese animals or nonalcoholic fatty liver disease (NAFLD) patients, let-7a and miR-34a levels were elevated with simultaneous decreases in ATG4B and Rab-8B levels. CONCLUSIONS: LXRα inhibits autophagy in hepatocytes through down-regulating ATG4B and Rab-8B by transcriptionally activating microRNA let-7a-2 and microRNA 34a genes and suppresses mitochondrial biogenesis and fuel consumption. This highlights a function of LXRα that culminates in the progression of liver steatosis and steatohepatitis, and the identified targets may be applied for a therapeutic strategy in the treatment of NAFLD.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/physiology , Cysteine Endopeptidases/metabolism , Hepatocytes/metabolism , Liver X Receptors/metabolism , Mitochondria/physiology , rab GTP-Binding Proteins/metabolism , Activation, Metabolic , Animals , Autophagy/genetics , Autophagy-Related Proteins/genetics , Cysteine Endopeptidases/genetics , Disease Models, Animal , Disease Progression , Down-Regulation , Fatty Liver/etiology , Fatty Liver/genetics , Fatty Liver/metabolism , Fatty Liver/physiopathology , Hep G2 Cells/metabolism , Hep G2 Cells/physiology , Hepatocytes/physiology , Humans , Lipid Metabolism/genetics , Lipid Metabolism/physiology , Liver/metabolism , Liver/physiology , Liver/physiopathology , Liver X Receptors/genetics , Liver X Receptors/physiology , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , MicroRNAs/physiology , Mitochondria/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Organelle Biogenesis , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Transcriptome , rab GTP-Binding Proteins/genetics
3.
Shock ; 53(4): 476-484, 2020 04.
Article in English | MEDLINE | ID: mdl-31232864

ABSTRACT

Severely burned patients suffer from a hypermetabolic syndrome that can last for years after the injury has resolved. The underlying cause of these metabolic alterations most likely involves the persistent elevated catecholamine levels that follow the surge induced by thermal injury. At the cellular level, endoplasmic reticulum (ER) stress in metabolic tissues is a hallmark observed in patients following burn injury and is associated with several detrimental effects. Therefore, ER stress could be the underlying cellular mechanism of persistent hypermetabolism in burned patients. Here, we show that catecholamines induce ER stress and that adreno-receptor blockers reduce stress responses in the HepG2 hepatocyte cell line. Our results also indicate that norepinephrine (NE) significantly induces ER stress in HepG2 cells and 3T3L1 mouse adipocytes. Furthermore, we demonstrate that the alpha-1 blocker, prazosin, and beta blocker, propranolol, block ER stress induced by NE. We also show that the effects of catecholamines in inducing ER stress are cell type-specific, as NE treatment failed to evoke ER stress in human fibroblasts. Thus, these findings reveal the mechanisms used by catecholamines to alter metabolism and suggest inhibition of the receptors utilized by these agents should be further explored as a potential target for the treatment of ER stress-mediated disease.


Subject(s)
Catecholamines/physiology , Endoplasmic Reticulum Stress/physiology , Fibroblasts/physiology , Hep G2 Cells/physiology , Receptors, Adrenergic, alpha/physiology , Receptors, Adrenergic, beta/physiology , Adrenergic alpha-1 Receptor Antagonists/pharmacology , Adrenergic beta-Antagonists/pharmacology , Cell Culture Techniques , Fibroblasts/drug effects , Hep G2 Cells/drug effects , Humans , Prazosin/pharmacology , Propranolol/pharmacology
4.
Biochem Biophys Res Commun ; 496(1): 238-243, 2018 01 29.
Article in English | MEDLINE | ID: mdl-29309789

ABSTRACT

To build a microfluidic device with various morphological features of the tumor vasculature for study of the effects of tumor vascular structures on the flow field and tumor cellular flow behaviors. The designed microfluidic device was able to approximatively simulate the in vivo structures of tumor vessels and the flow within it. In this models, the influences of the angle of bifurcation, the number of branches, and the narrow channels on the flow field and the influence of vorticity on the retention of HepG2 cells were significant. Additionally, shear stress below physiological conditions of blood circulation has considerable effect on the formation of the lumen-like structures (LLSs) of HepG2 cells. These results can provide some data and reference in the understanding of the interaction between hemorheological properties and tumor vascular structures in solid tumors.


Subject(s)
Bioreactors , Cell Culture Techniques/instrumentation , Cell Separation/instrumentation , Hep G2 Cells/physiology , Lab-On-A-Chip Devices , Neovascularization, Pathologic/physiopathology , Equipment Design , Equipment Failure Analysis , Hep G2 Cells/pathology , Humans , Neovascularization, Pathologic/pathology
5.
Int J Obes (Lond) ; 41(9): 1403-1412, 2017 09.
Article in English | MEDLINE | ID: mdl-28559541

ABSTRACT

Background &aims:Low levels of adiponectin (APN), an adipose-derived adipokine, are associated with obesity and non-alcoholic steatohepatitis although its role in high-fat diet-induced hepatic injury and steatosis remains unclear. Here we hypothesized that APN deficiency alters fat diet-induced hepatic function. To this end, we examined the effect of APN deficiency on high-fat diet-induced hepatic injury, apoptosis and steatosis. METHODS: Adult wild type and APN knockout mice were fed a low- or high-fat diet for 20 weeks. Serum levels of liver enzymes aspartate aminotransferase (AST), alanine aminotransferase (ALT), cholesterol, hepatic triglycerides, steatosis, pro-inflammatory cytokines, apoptosis and autophagy were examined. RESULTS: High-fat feeding led to elevated body (48.2%) and liver weights (18.8%), increased levels of ALT (87.8%), serum cholesterol (104.4%), hepatic triglycerides (305.6%) and hepatic fat deposition as evidenced by Oil Red O staining, along with a reduced AST/ALT ratio and unchanged AST. Although APN knockout itself did not affect hepatic function and morphology, it reconciled fat diet-induced hepatic injury (P<0.05 vs WT-HF group) without reversing changes in body and liver weights, serum cholesterol and hepatic steatosis. In addition, fat diet intake promoted AMPK phosphorylation, p62 accumulation and apoptosis, including elevated Bax and cleaved Caspase-3 and downregulated Bcl-2, along with suppressed phosphorylation of Akt, STAT3 and JNK, and the autophagy makers Atg7, Beclin-1 and LC3B (P<0.05 vs WT-LF group) without affecting hepatic interlelukin-6 and tumor necrosis factor-α levels, the effects were reversed or significantly attenuated by APN knockout (P<0.05 vs WT-HF group). In vitro study using HepG2 cells revealed that STAT3 activation rescued palmitic acid-induced cell injury whereas STAT3 inhibition nullified APN knockdown-offered beneficial effects. CONCLUSIONS: Our results revealed that high-fat diet intake promotes hepatic steatosis, apoptosis and interrupted autophagy. APN knockout elicits protective effect against hepatic injury possibly associated with autophagy regulation despite persistent hepatic steatosis.


Subject(s)
Adiponectin/deficiency , Apoptosis/physiology , Autophagy/physiology , Diet, High-Fat/adverse effects , Hep G2 Cells/physiology , Liver/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Alanine Transaminase/metabolism , Animals , Blotting, Western , Cells, Cultured , Disease Models, Animal , Humans , Lipid Metabolism , Male , Mice , Mice, Knockout , STAT3 Transcription Factor/physiology , Signal Transduction , Triglycerides/metabolism
6.
Zhejiang Da Xue Xue Bao Yi Xue Ban ; 45(4): 403-409, 2016 05 25.
Article in Chinese | MEDLINE | ID: mdl-27868414

ABSTRACT

Objective: To observe the effect of uniform and shift rotation culture on the formation and activity of the alginate-chitosan (AC) microencapsulated HepLL immortalized human hepatocytes and HepG2 cells aggregates. Methods: AC microcapsulated HepG2 and HepLL cells were randomly divided into two groups. Each group was divided into 3 subgroups according to uniform and shift rotation culture.The size and number of aggregates were observed and measured under laser confocal microscopy and inverted microscope dynamically. The amount of albumin synthesis was detected by ELISA, the clearance of ammonia was detected by colorimetry, and diazepam conversion function was detected by high performance liquid chromatography (HPLC). Results: On day 6, 8, 10, 12, 14 and 16, the number and size of the aggregates, albumin synthesis, diazepam clearance and ammonium clearance increased significantly in shift rotation culture group than in uniform group (all P<0.01). The albumin synthesis, diazepam clearance, and ammonium clearance in the microencapsulated HepLL groups were significantly higher than those of HepG2 cells at any time (all P<0.01). Conclusion: Shift rotation culture can significantly promote the formation and increase the activity of AC microencapsulated HepLL and HepG2 aggregates, and HepLL cells may be more suitable for bioartificial liver than HepG2.


Subject(s)
Cell Aggregation/physiology , Cell Culture Techniques/methods , Hep G2 Cells/physiology , Hepatocytes/physiology , Albumins/biosynthesis , Albumins/metabolism , Alginates , Ammonia/metabolism , Animals , Cell Line, Transformed/physiology , Chitosan , Diazepam/metabolism , Glucuronic Acid , Hep G2 Cells/cytology , Hepatocytes/cytology , Hexuronic Acids , Humans , Liver, Artificial , Rotation
7.
Zhonghua Zhong Liu Za Zhi ; 38(10): 725-730, 2016 Oct 23.
Article in Chinese | MEDLINE | ID: mdl-27784453

ABSTRACT

Objective: To investigate the killing effect of low-temperature plasma (LTP) on HepG2, A549 and HeLa cell lines and explore its possible mechanism. Methods: The inhibitory effect of LTP on the proliferation of HepG2, A549 and HeLa cells was determined by MTT assay. Transmission electron microscopy was used to observe the ultrastructural changes of HepG2, A549 and HeLa cells treated with LTP. Cell apoptosis was detected by Muse cytometry. Western blot was used to detect the expression of apoptosis-related proteins. Results: The survival rates of LTP-irradiated HepG2 cells (irradiated for 107 s), HeLa cells (irradiated for 121 s) and A549 cells (irradiated for 127 s) were 50%. LTP destroyed the ultrastructure of HepG2, A549 and HeLa cells to different degrees, showing nuclear fragmentation and organelle damages. The apoptosis rates of the three cell lines were increased at 24 h after exposure to LTP for 1/6 IC50 irradiation time. Furthermore, LTP irradiation also suppressed the protein expression of Bcl-2 and XRCC1 and increased that of Bax. Conclusions: LTP has an obvious killing effect on HepG2, A549 and HeLa cancer cell lines. This effect may be related to the induction of cell apoptosis and inhibition of DNA repair.


Subject(s)
A549 Cells/physiology , Apoptosis , Cell Proliferation , Cryotherapy/methods , HeLa Cells/physiology , Hep G2 Cells/physiology , A549 Cells/radiation effects , A549 Cells/ultrastructure , Apoptosis Regulatory Proteins/metabolism , Cell Proliferation/radiation effects , Cell Survival/radiation effects , HeLa Cells/radiation effects , HeLa Cells/ultrastructure , Hep G2 Cells/radiation effects , Hep G2 Cells/ultrastructure , Humans
8.
Dig Dis Sci ; 61(4): 1098-106, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26668056

ABSTRACT

BACKGROUND: Although hepatocellular carcinoma cells can sometimes undergo differentiation in an embryonic microenvironment, the mechanism is poorly understood. AIM: The developmental stage-specific embryonic induction of tumor cell differentiation was investigated. METHODS: Both chick and mouse liver extracts and hepatoblast-enriched cells at different developmental stages were used to treat human hepatoma HepG2 cells, and the effects on the induction of differentiation were evaluated. The nuclear factors controlling differentiation, hepatocyte nuclear factor (HNF)-4α, HNF-1α, HNF-6 and upstream stimulatory factor-1 (USF-1), and the oncogene Myc and alpha-fetoprotein (AFP) were measured. HNF-4α RNA interference was used to verify the role of HNF-4α. Embryonic induction effects were further tested in vivo by injecting HepG2 tumor cells into immunodeficient nude mice. RESULTS: The 9-11-days chick liver extracts and 13.5-14.5-days mouse hepatoblast-enriched cells could inhibit proliferation and induce differentiation of HepG2 cells, leading to either death or maturation to hepatocytes. The maturation of surviving HepG2 cells was confirmed by increases in the expressions of HNF-4α, HNF-1α, HNF-6, and USF-1, and decreases in Myc and AFP. The embryonic induction of HepG2 cell maturation could be attenuated by HNF-4α RNA interference. Furthermore, the 13.5-days mouse hepatoblast culture completely eliminated HepG2 tumors with inhibited Myc and induced HNF-4α, confirming this embryonic induction effect in vivo. CONCLUSIONS: This study demonstrated that developmental stage-specific embryonic induction of HepG2 cell differentiation might help in understanding embryonic differentiation and oncogenesis.


Subject(s)
Embryonic Induction , Hep G2 Cells/physiology , Animals , Chick Embryo , Hepatocyte Nuclear Factor 4/metabolism , Humans , Liver Extracts , Mice
9.
Appl Microbiol Biotechnol ; 100(6): 2677-84, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26536876

ABSTRACT

Staphylococcal enterotoxins C2 (SEC2) is a classical model of superantigens (SAg), which has the powerful ability to activate T cells as well as induce massive cytokine production. This property makes SEC2 and its mutants well concerned as a potential new immune-regulatory agent for cancer therapy. We previously constructed a SEC2 mutant named SAM-3, which had prominently antitumor activity in BALB/c mice model. But, the underlying molecular mechanism for stimulation of human peripheral blood mononuclear cells (PBMCs) and antitumor effect on human tumor cells induced by SAM-3 is not clear. Here, we showed that SAM-3 could activate human TCR Vß 12, 13A, 14, 15, 17, and 20 CD8(+) subgroup T cells, which secreted the cytokines IL-2, IFN-γ, and TNF-α, and exhibit stimulation activity in a dose-dependent manner. TNF-α secreted from activated T cells could induce apoptosis and G1-phase arrest and lead to the antitumor effect in HepG2 cells. Meanwhile, SAM-3 upregulated the expression of tumor necrosis factor receptor 1 (TNFR1) mRNA and activity of caspase-3 and caspase-8. We also found that the antitumor activity and activity of caspase-3 and caspase-8 were decreased when the neutralizing TNF-α monoclonal antibody presented. These data suggest that TNF-α secreted by SAM-3-activated T cells is an important factor in inducing apoptosis in HepG2 cells.


Subject(s)
Antineoplastic Agents/metabolism , Apoptosis , Enterotoxins/metabolism , Hep G2 Cells/physiology , Mutant Proteins/metabolism , T-Lymphocytes/physiology , Tumor Necrosis Factor-alpha/metabolism , Enterotoxins/genetics , Hep G2 Cells/drug effects , Humans , Lymphocyte Activation , Mutant Proteins/genetics , T-Lymphocytes/drug effects
10.
Med Hypotheses ; 85(6): 736-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26519102

ABSTRACT

Alcoholic liver disease is a major source of morbidity and mortality worldwide. Twin studies had demonstrated heritability of alcoholic liver disease. Although to date only Adiponutrin (PNPLA3) rs738409 polymorphism (I148M) had been unequivocally proved to be associated with increased risk of alcoholic liver disease across different ethnicities. This protein was previously thought to have a predominant lipolytic role. However, recent investigations have provided evidence of lipogenic activity of this protein. The current hypothesis paper is summarizing the recent evidences gleaned in biological role of Adiponutrin and bioinformatic pointers towards a role in lipid trafficking. A critical appraisal of the utility of murine models and cell based systems in investigating Adiponutrin is also presented. As the HepG2 cell line harbors the I148M mutation in homozygous state it is hypothesized that this should represent an ideal model system for PNPLA3 biology. Thus, as Adiponutrin is proposed as having both lipolytic and lipogenic/lipid trafficking roles it is termed as a Yin-Yang protein in analogy to ancient Chinese wisdom.


Subject(s)
Disease Models, Animal , Hep G2 Cells/physiology , Lipid Metabolism/genetics , Liver Cirrhosis/genetics , Liver Diseases, Alcoholic/genetics , Phospholipases A2, Calcium-Independent/genetics , Animals , Genetic Predisposition to Disease/genetics , Hep G2 Cells/pathology , Humans , Liver Cirrhosis/pathology , Liver Diseases, Alcoholic/metabolism , Mice , Models, Genetic , Phospholipases A2, Calcium-Independent/metabolism
11.
Biosci Rep ; 35(3)2015 May 07.
Article in English | MEDLINE | ID: mdl-26182370

ABSTRACT

3D (three-dimensional) cultures are considered to be an effective method for toxicological studies; however, little evidence has been reported whether 3D cultures have an impact on hepatocellular physiology regarding lipid or glucose metabolism. In the present study, we conducted physiological characterization of hepatoma cell lines HepG2 and HepaRG cells cultured in 3D conditions using a hanging drop method to verify the effect of culture environment on cellular responses. Apo (Apolipoprotein)B as well as albumin secretion was augmented by 3D cultures. Expression of genes related to not only drug, but also glucose and lipid metabolism were significantly enhanced in 3D cultured HepaRG spheroids. Furthermore, mRNA levels of CYP (cytochrome P450) enzymes following exposure to corresponding inducers increased under the 3D condition. These data suggest that this simple 3D culture system without any special biomaterials can improve liver-specific characteristics including lipid metabolism. Considering that the system enables high-throughput assay, it may become a powerful tool for compound screening concerning hepatocellular responses in order to identify potential drugs.


Subject(s)
Cell Culture Techniques/methods , Liver/physiology , Spheroids, Cellular , Transcriptome , Albumins/metabolism , Apolipoproteins B/metabolism , Cell Culture Techniques/instrumentation , Cytochrome P-450 Enzyme Inducers/pharmacology , Cytochrome P-450 Enzyme System/genetics , Glucose/genetics , Glucose/metabolism , Hep G2 Cells/drug effects , Hep G2 Cells/physiology , High-Throughput Screening Assays/methods , Humans , Lipid Metabolism/genetics , Lipoproteins, LDL/genetics , Lipoproteins, LDL/metabolism , Liver/metabolism , Omeprazole/pharmacology , Phenobarbital/pharmacology , Rifampin/pharmacology
12.
J Appl Toxicol ; 35(7): 709-16, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25042557

ABSTRACT

Autophagy plays an important role in response to intracellular and extracellular stress to sustain cell survival. However, dysregulated or excessive autophagy may lead to cell death, known as "type II programmed cell death," and it is closely associated with apoptosis. In our previous study, we proposed that olaquindox induced apoptosis of HepG2 cells through a caspase-9 dependent mitochondrial pathway. In this study, we investigated autophagy induced by olaquindox and explored the crosstalk between apoptosis and autophagy in olaquindox-treated HepG2 cells. Olaquindox-induced autophagy was demonstrated by the accumulation of monodansylcadervarine, as well as elevated expression of autophagy-related MAP-LC3 and Beclin 1 proteins. The autophagy inhibitor 3-methyladenine significantly increased the apoptotic rate induced by olaquindox, which was correlated with increased ratio of Bax/Bcl-2. The further studies showed that olaquindox increased the levels of reactive oxygen species (ROS), and antioxidant N-acetyl-L-cysteine (NAC) effectively blocked the accumulation of ROS but failed to block autophagy. Moreover, olaquindox induced the activation of c-Jun N-terminal protein kinase (JNK), and JNK inhibitor SP600125 failed to block autophagy. Instead, olaquindox-induced autophagy was enhanced by NAC or SP600125. Meanwhile, JNK activation was remarkably blocked by NAC, indicating that ROS may be the upstream signaling molecules of JNK activation and involved in the negative regulation of olaquindox-induced autophagy. These results suggest that olaquindox induces autophagy in HepG2 cells and that olaquindox-induced apoptosis can be enhanced by 3-methyladenine. Olaquindox-induced autophagy in HepG2 cells is upregulated by Beclin 1 but downregulated by ROS-dependent JNK.


Subject(s)
Autophagy/drug effects , Hep G2 Cells/drug effects , JNK Mitogen-Activated Protein Kinases/drug effects , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Anthracenes/pharmacology , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Autophagy/physiology , Beclin-1 , Cadaverine/analogs & derivatives , Cadaverine/metabolism , Down-Regulation/drug effects , Hep G2 Cells/physiology , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/physiology , Membrane Proteins/metabolism
13.
Integr Biol (Camb) ; 6(6): 596-602, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24801132

ABSTRACT

We report a biocompatible method of selectively retrieving 3-D cell-encapsulating hydrogel microstructures from a culture substrate. First, poly(l-lysine)/hyaluronic acid (PLL/HA) polyelectrolyte multilayers (PEMs) with methacrylated chitosan (GMA-Chi) on top were formed on an ITO substrate. Then, a cell-encapsulating hydrogel micropattern was formed; a HepG2 cell-encapsulating heparin-based hydrogel micropattern was fabricated by thiol-ene photopolymerization. The application of an oxidative potential of 2 V resulted in the detachment of the cell-encapsulating hydrogels by the dissolution of PEMs. The time of complete retrieval of the hydrogels was controllable by modulating the number of PEM layers. The applied potential did not affect the viability or the function of the cells in the entire hydrogels. In contrast, when a reductive electrochemical potential (-1.8 V) was applied to a silane-modified ITO to release cell-encapsulating hydrogels by the desorption of silane [Chem. Commun., 2009, 5865], extensive cell death at the bottom of the hydrogel adjacent to the electrode was observed.


Subject(s)
Chitosan/chemistry , Hep G2 Cells/physiology , Hyaluronic Acid/chemistry , Hydrogels/pharmacology , Polylysine/chemistry , Cell Survival , Electrochemical Techniques , Humans , Microscopy, Confocal
14.
Biochem Biophys Res Commun ; 449(1): 175-81, 2014 Jun 20.
Article in English | MEDLINE | ID: mdl-24832736

ABSTRACT

Much of the difficulty in elucidating the precise function of S100 protein family has been attributed to functional redundancy and compensation by its conserved family members. In this study, we showed that seven S100 family members were almost totally undetectable in HepG2.2.15 cells, while all of them were highly expressed in its parental HepG2 cells. Re-expression of S100 proteins in HepG2.2.15 cells can partially rescue their defects in cell protrusion and migration through the regulation of cytoskeletons and adhesions. Thus, HepG2.2.15 can serve as a useful model for studying cell protrusion and migration regulated by S100 proteins.


Subject(s)
Cell Enlargement , Cell Movement/physiology , Hep G2 Cells/pathology , Hep G2 Cells/physiology , S100 Proteins/physiology , Hep G2 Cells/classification , Humans
15.
Int J Biol Sci ; 10(5): 490-9, 2014.
Article in English | MEDLINE | ID: mdl-24795530

ABSTRACT

Though gallotannin was known to have anti-oxidant and antitumor activity, the underlying antitumor mechanism of gallotannin still remains unclear. Thus, in the present study, antitumor mechanism of gallotannin was elucidated in hepatocellular carcinoma cells. Gallotannin significantly exerted cytotoxicity against Hep G2 and Chang hepatocellular carcinoma cells with the accumulation of the sub-G1 population and increase of terminal deoxynucleotidyltransferasedUTP nick end labeling (TUNEL) positive cells as an apoptotic feature. Also, gallotannin attenuated the expression of pro-caspase9, pro-caspase3, Bcl2 and integrin ß1 and cleaved poly(ADP)-ribose polymerase (PARP) in Hep G2 and Chang cancer cells. Furthermore, gallotannin suppressed cell repair motility by wound healing assay and also inhibited cell adhesion in Hep G2 cells. Of note, gallotannin attenuated the expression of epithelial cadherin (E-cadherin) to form cell-cell adhesion from the early stage, and also beta-catenin at late phase in Hep G2 cells. Consistently, Immunofluorescence assay showed that E-cadherin or ß-catenin expression was suppressed in a time dependent manner by gallotannin. Furthermore, silencing of E-cadherin by siRNA transfection method enhanced PAPR cleavage, caspase 3 activation and sub G1 population and attenuated the cell adhesion induced by gallotannin in Hep G2 cells. Overall, our findings demonstrate that the disruption of cell adhesion junction by suppression of E-cadherin mediates gallotannin enhanced apoptosis in Hep G2 liver cancer cells.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/physiology , Cadherins/metabolism , Carcinoma, Hepatocellular/physiopathology , Hep G2 Cells/physiology , Hydrolyzable Tannins/pharmacology , Liver Neoplasms/physiopathology , Apoptosis/drug effects , Carcinoma, Hepatocellular/metabolism , Fluorescent Antibody Technique , Gene Expression Regulation, Neoplastic/drug effects , Hep G2 Cells/metabolism , Humans , In Situ Nick-End Labeling , Liver Neoplasms/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA Interference , RNA, Small Interfering/genetics
16.
J Formos Med Assoc ; 113(10): 680-7, 2014 Oct.
Article in English | MEDLINE | ID: mdl-23849456

ABSTRACT

BACKGROUND/PURPOSE: Using gradient ionic detergent, we optimized the preparation procedure for the decellularized liver biologic scaffold, and analyzed its immunogenicity and biocompatibility. METHODS: EDTA, hypotonic alkaline solution, Triton X-100, and gradient sodium dodecyl sulfate (1%, 0.5%, and 0.1%, respectively) were prepared for continuous perfusion through the hepatic vascular system. The decellularization of the liver tissue was performed with the optimized reagent buffer and washing protocol. In addition, the preservation of the original extracellular matrix was observed. To analyze its biocompatibility, the scaffold was embedded in a heterologous animal and the inflammation features, including the surrounding cell infiltration and changes of the scaffold architecture, were detected. The cell-attachment ability was also validated by the perfusion culture of HepG2 cells with the scaffold. RESULTS: By using gradient ionic detergent, we completed the decellularization process in approximately 5 h, which was shorter than >10 hours in previous experiments (p<0.001). The extracellular matrix was kept relatively intact, with no obvious inflammatory cellular infiltration or structural damage in the grafted tissue. The engraftment efficiencies of HepG2 were 86±5% (n=8). The levels of albumin and urea synthesis were significantly superior to the ones in traditional two-dimensional culture. CONCLUSION: The current new method can be used efficiently for the decellularization of the liver biologic scaffold with satisfying biocomparability for application both in vivo and in vitro.


Subject(s)
Cell Culture Techniques/methods , Extracellular Matrix/transplantation , Liver/cytology , Tissue Engineering/methods , Tissue Scaffolds , Albumins/biosynthesis , Animals , Blood Vessel Prosthesis , Female , Hep G2 Cells/physiology , Humans , Male , Matrix Attachment Regions/physiology , Perfusion , Rabbits , Rats , Rats, Sprague-Dawley , Urea/analysis
17.
PLoS Biol ; 11(12): e1001739, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24358023

ABSTRACT

The development and maintenance of polarized epithelial tissue requires a tightly controlled orientation of mitotic cell division relative to the apical polarity axis. Hepatocytes display a unique polarized architecture. We demonstrate that mitotic hepatocytes asymmetrically segregate their apical plasma membrane domain to the nascent daughter cells. The non-polarized nascent daughter cell can form a de novo apical domain with its new neighbor. This asymmetric segregation of apical domains is facilitated by a geometrically distinct "apicolateral" subdomain of the lateral surface present in hepatocytes. The polarity protein partitioning-defective 1/microtubule-affinity regulating kinase 2 (Par1b/MARK2) translates this positional landmark to cortical polarity by promoting the apicolateral accumulation of Leu-Gly-Asn repeat-enriched protein (LGN) and the capture of nuclear mitotic apparatus protein (NuMA)-positive astral microtubules to orientate the mitotic spindle. Proliferating hepatocytes thus display an asymmetric inheritance of their apical domains via a mechanism that involves Par1b and LGN, which we postulate serves the unique tissue architecture of the developing liver parenchyma.


Subject(s)
Cell Membrane/physiology , Cell Polarity/physiology , Hepatocytes/physiology , Intracellular Signaling Peptides and Proteins/physiology , Metalloproteases/physiology , Mitochondrial Proteins/physiology , Spindle Apparatus/physiology , Cell Proliferation , Hep G2 Cells/physiology , Humans
18.
Cell Struct Funct ; 38(1): 109-21, 2013.
Article in English | MEDLINE | ID: mdl-23575155

ABSTRACT

Human mesenchymal stem cells (hMSCs) accumulate at carcinomas and have a great impact on cancer cell's behavior. Here we demonstrated that hMSCs could display both the promotional and inhibitive effects on growth of HepG2 and Hela cells by using the conditioned media, indirect co-culture, and cell-to-cell co-culture. Cell growth was increased following the addition of lower proportion of hMSCs while decreased by treatment of higher proportion of hMSCs. We also established a novel noninvasive label way by using internalizing quantum dots (i-QDs) for study of cell-cell contact in the co-culture, which was effective and sensitive for both tracking and distinguishing different cells population without the disturbance of cells. Furthermore, we investigated the role of hMSCs in regulation of cell growth and showed that mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) signaling pathways were involved in hMSC-mediated cell inhibition and proliferation. Our findings suggested that hMSCs regulated cancer cell function by providing a suitable environment, and the discovery from the study would provide some clues for development of effective strategy for hMSC-based cancer therapies.


Subject(s)
Cell Communication/physiology , Cell Proliferation , HeLa Cells/pathology , Hep G2 Cells/pathology , Mesenchymal Stem Cells/cytology , Cell Communication/drug effects , Cell Proliferation/drug effects , Coculture Techniques , Culture Media, Conditioned/pharmacology , Female , HeLa Cells/drug effects , HeLa Cells/physiology , Hep G2 Cells/drug effects , Hep G2 Cells/physiology , Humans , Liver Neoplasms/pathology , Liver Neoplasms/physiopathology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/physiology , Mitogen-Activated Protein Kinase Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/physiopathology
19.
Hepatobiliary Pancreat Dis Int ; 11(6): 594-605, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23232630

ABSTRACT

BACKGROUND: Orthotopic liver transplantation (OLT) is the most effective therapy for liver failure. However, OLT is severely limited by the shortage of liver donors. Bioartificial liver (BAL) shows great potential as an alternative therapy for liver failure. In recent years, progress has been made in BAL regarding genetically engineered cell lines, immortalized human hepatocytes, methods for preserving the phenotype of primary human hepatocytes, and other functional hepatocytes derived from stem cells. DATA SOURCES: A systematic search of PubMed and ISI Web of Science was performed to identify relevant studies in English language literature using the key words such as liver failure, bioartificial liver, hepatocyte, stem cells, differentiation, and immortalization. More than 200 articles related to the cell sources of hepatocyte in BAL were systematically reviewed. RESULTS: Methods for preserving the phenotype of primary human hepatocytes have been successfully developed. Many genetically engineered cell lines and immortalized human hepatocytes have also been established. Among these cell lines, the incorporation of BAL with GS-HepG2 cells or alginate-encapsulated HepG2 cells could prolong the survival time and improve pathophysiological parameters in an animal model of liver failure. The cBAL111 cells were evaluated using the AMC-BAL bioreactor, which could eliminate ammonia and lidocaine, and produce albumin. Importantly, BAL loading with HepLi-4 cells could significantly improve the blood biochemical parameters, and prolong the survival time in pigs with liver failure. Other functional hepatocytes differentiated from stem cells, such as human liver progenitor cells, have been successfully achieved. CONCLUSIONS: Aside from genetically modified liver cell lines and immortalized human hepatocytes, other functional hepatocytes derived from stem cells show great potential as cell sources for BAL. BAL with safe and effective liver cells may be achieved for clinical liver failure in the near future.


Subject(s)
Hep G2 Cells/physiology , Hepatocytes/physiology , Liver, Artificial , Stem Cells/physiology , Animals , Cell Culture Techniques , Genetic Engineering , Humans , Swine
20.
World J Gastroenterol ; 18(13): 1485-95, 2012 Apr 07.
Article in English | MEDLINE | ID: mdl-22509080

ABSTRACT

AIM: To investigate the possible mechanism by which hepatitis B virus X protein (HBx) mediates apoptosis of HepG2 cells. METHODS: HBx expression vector pcDNA3.1-X was transfected into HepG2 cells to establish an HBx high-expression cellular model as pcDNA3.1-X transfected group. The pcDNA3.1-X and pSilencer3.1-shHBX (HBx antagonist) were cotransfected into HepG2 cells to establish an HBx low-expression model as RNAi group. Untransfected HepG2 cells and HepG2 cells transfected with negative control plasmid were used as controls. Apoptosis rate, the expression of Fas/FasL signaling pathway-related proteins and the phosphorylation levels of MLK3, MKK7 and JNKs, which are upstream molecules of death receptor pathways and belong to the family of mitogen-activated protein kinases (MAPKs), were measured in each group. RESULTS: Compared with HepG2 cell group and RNAi group, apoptosis rate, the expression of Fas and FasL proteins, and the activation of MLK3, MKK7 and JNKs were increased in the pcDNA3.1-X transfected group. The activation of JNKs and expression of FasL protein were inhibited in the pcDNA3.1-X transfected group when treated with a known JNK inhibitor, SP600125. When authors treated pcDNA3.1-X transfected group with K252a, a known MLK3 inhibitor, the activation of MLK3, MKK7 and JNKs as well as expression of FasL protein was inhibited. Furthermore, cell apoptosis rate was also significantly declined in the presence of K252a in the pcDNA3.1-X transfected group. CONCLUSION: HBx can induce HepG2 cell apoptosis via a novel active MLK3-MKK7-JNKs signaling module to upregulate FasL protein expression.


Subject(s)
Apoptosis/physiology , Fas Ligand Protein/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase 7/metabolism , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/physiology , Trans-Activators/metabolism , Animals , Caspases/metabolism , Enzyme Activation , Enzyme Inhibitors/metabolism , Hep G2 Cells/physiology , Humans , JNK Mitogen-Activated Protein Kinases/genetics , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase Kinases/genetics , Trans-Activators/genetics , Viral Regulatory and Accessory Proteins , fas Receptor/metabolism , Mitogen-Activated Protein Kinase Kinase Kinase 11
SELECTION OF CITATIONS
SEARCH DETAIL
...