Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Pharm Anal ; 13(3): 315-322, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37102107

ABSTRACT

Trace amines (TAs) are metabolically related to catecholamine and associated with cancer and neurological disorders. Comprehensive measurement of TAs is essential for understanding pathological processes and providing proper drug intervention. However, the trace amounts and chemical instability of TAs challenge quantification. Here, diisopropyl phosphite coupled with chip two-dimensional (2D) liquid chromatography tandem triple-quadrupole mass spectrometry (LC-QQQ/MS) was developed to simultaneously determine TAs and associated metabolites. The results showed that the sensitivities of TAs increased up to 5520 times compared with those using nonderivatized LC-QQQ/MS. This sensitive method was utilized to investigate their alterations in hepatoma cells after treatment with sorafenib. The significantly altered TAs and associated metabolites suggested that phenylalanine and tyrosine metabolic pathways were related to sorafenib treatment in Hep3B cells. This sensitive method has great potential to elucidate the mechanism and diagnose diseases considering that an increasing number of physiological functions of TAs have been discovered in recent decades.

2.
Nat Commun ; 13(1): 4291, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35879296

ABSTRACT

Whether amino acids act on cellular insulin signaling remains unclear, given that increased circulating amino acid levels are associated with the onset of type 2 diabetes (T2D). Here, we report that phenylalanine modifies insulin receptor beta (IRß) and inactivates insulin signaling and glucose uptake. Mice fed phenylalanine-rich chow or phenylalanine-producing aspartame or overexpressing human phenylalanyl-tRNA synthetase (hFARS) develop insulin resistance and T2D symptoms. Mechanistically, FARS phenylalanylate lysine 1057/1079 of IRß (F-K1057/1079), inactivating IRß and preventing insulin from promoting glucose uptake by cells. SIRT1 reverse F-K1057/1079 and counteract the insulin-inactivating effects of hFARS and phenylalanine. F-K1057/1079 and SIRT1 levels in white blood cells from T2D patients are positively and negatively correlated with T2D onset, respectively. Blocking F-K1057/1079 with phenylalaninol sensitizes insulin signaling and relieves T2D symptoms in hFARS-transgenic and db/db mice. These findings shed light on the activation of insulin signaling and T2D progression through inhibition of phenylalanylation.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Animals , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Humans , Insulin , Insulin Resistance/physiology , Mice , Phenylalanine , Sirtuin 1/genetics
3.
Adv Sci (Weinh) ; 9(20): e2103887, 2022 07.
Article in English | MEDLINE | ID: mdl-35187863

ABSTRACT

Cancer cells are addicted to glutamine. However, cancer cells often suffer from glutamine starvation, which largely results from the fast growth of cancer cells and the insufficient vascularization in the interior of cancer tissues. Herein, based on clinical samples, patient-derived cells (PDCs), and cell lines, it is found that liver cancer cells display stem-like characteristics upon glutamine shortage due to maintaining the stemness of tumor initiating cells (TICs) and even promoting transformation of non-TICs into stem-like cells by glutamine starvation. Increased expression of glutamine synthetase (GS) is essential for maintaining and promoting stem-like characteristics of liver cancer cells during glutamine starvation. Mechanistically, glutamine starvation activates Rictor/mTORC2 to induce HDAC3-mediated deacetylation and stabilization of GS. Rictor is significantly correlated with the expression of GS and stem marker OCT4 at tumor site, and closely correlates with poor prognosis of hepatocellular carcinomas. Inhibiting components of mTORC2-HDAC3-GS axis decrease TICs and promote xenografts regression upon glutamine-starvation therapy. Collectively, the data provides novel insights into the role of Rictor/mTORC2-HDAC3 in reprogramming glutamine metabolism to sustain stemness of cancer cells. Targeting Rictor/HDAC3 may enhance the efficacy of glutamine-starvation therapy and limit the rapid growth and malignant progression of tumors.


Subject(s)
Liver Neoplasms , Cell Line , Glutamate-Ammonia Ligase , Glutamine/deficiency , Glutamine/metabolism , Histone Deacetylases , Humans , Mechanistic Target of Rapamycin Complex 2/metabolism , Transcription Factors
4.
Oncogenesis ; 10(11): 74, 2021 Nov 12.
Article in English | MEDLINE | ID: mdl-34772914

ABSTRACT

Aberrant lipid metabolism is an essential feature of hepatocellular carcinoma (HCC). Fatty acid transport protein-5 (FATP5) is highly expressed in the liver and is involved in the fatty acid transport pathway. However, the potential role of FATP5 in the pathogenesis of HCC remains largely unknown. Herein, we showed that FATP5 was downregulated in HCC tissues and even much lower in vascular tumor thrombi. Low expression of FATP5 was correlated with multiple aggressive and invasive clinicopathological characteristics and contributed to tumor metastasis and a poor prognosis in HCC patients. FATP5 inhibited the epithelial-mesenchymal transition (EMT) process and suppressed HCC cell migration and invasion, while silencing FATP5 had the opposite effects. Mechanistically, knockdown of FATP5 promoted cellular glycolytic flux and ATP production, thus suppressing AMP-activated protein kinase (AMPK) and activating its downstream signaling mammalian target of rapamycin (mTOR) to support HCC progression and metastasis. Activation of AMPK using metformin reversed the EMT program and impaired the metastatic capacity of FATP5-depleted HCC cells. Collectively, FATP5 served as a novel suppressor of HCC progression and metastasis partly by regulating the AMPK/mTOR pathway in HCC, and targeting the FATP5-AMPK axis may be a promising therapeutic strategy for personalized HCC treatment.

5.
Cancer Lett ; 420: 26-37, 2018 04 28.
Article in English | MEDLINE | ID: mdl-29409972

ABSTRACT

Obesity is a major risk factor for hepatocellular carcinoma (HCC) and is typically accompanied by higher levels of serum dipeptidyl peptidase 4 (DPP4). However, the role of DPP4 in obesity-promoted HCC is unclear. Here, we found that consumption of a high-fat diet (HFD) promoted HCC cell proliferation and metastasis and led to poor survival in a carcinogen-induced model of HCC in rats. Notably, genetic ablation of DPP4 or treatment with a DPP4 inhibitor (vildagliptin) prevented HFD-induced HCC. Moreover, HFD-induced DPP4 activity facilitated angiogenesis and cancer cell metastasis in vitro and in vivo, and vildagliptin prevented tumor progression by mediating the pro-angiogenic role of chemokine ligand 2 (CCL2). Loss of DPP4 effectively reversed HFD-induced CCL2 production and angiogenesis, indicating that the DPP4/CCL2/angiogenesis cascade had key roles in HFD-associated HCC progression. Furthermore, concomitant changes in serum DPP4 and CCL2 were observed in 210 patients with HCC, and high serum DPP4 activity was associated with poor clinical prognosis. These results revealed a link between obesity-related high serum DPP4 activity and HCC progression. Inhibition of DPP4 may represent a novel therapeutic intervention for patients with HCC.


Subject(s)
Carcinoma, Hepatocellular/pathology , Chemokine CCL2/metabolism , Diet, High-Fat/adverse effects , Dipeptidyl Peptidase 4/blood , Dipeptidyl Peptidase 4/genetics , Liver Neoplasms/pathology , Obesity/complications , Animals , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/prevention & control , Cell Line, Tumor , Cell Proliferation , Disease Progression , Down-Regulation , Gene Expression Regulation, Neoplastic/drug effects , Humans , Liver Neoplasms/blood supply , Liver Neoplasms/etiology , Liver Neoplasms/prevention & control , Male , Mice , Neoplasm Metastasis , Neoplasm Transplantation , Obesity/blood , Obesity/chemically induced , Obesity/drug therapy , Prognosis , Rats , Vildagliptin/administration & dosage , Vildagliptin/pharmacology
6.
Hepatology ; 65(5): 1628-1644, 2017 05.
Article in English | MEDLINE | ID: mdl-28027570

ABSTRACT

Potential biomarkers that can be used to determine prognosis and perform targeted therapies are urgently needed to treat patients with hepatocellular carcinoma (HCC). To meet this need, we performed a screen to identify functional genes associated with hepatocellular carcinogenesis and its progression at the transcriptome and proteome levels. We identified aldehyde dedydrogenase-2 (ALDH2) as a gene of interest for further study. ALDH2 levels were significantly lower at the mRNA and protein level in tumor tissues than in normal tissues, and they were even lower in tissues that exhibited increased migratory capacity. A study of clinical associations showed that ALDH2 is correlated with survival and multiple migration-associated clinicopathological traits, including the presence of metastasis and portal vein tumor thrombus. The result of overexpressing or knocking down ALDH2 showed that this gene inhibited migration and invasion both in vivo and in vitro. We also found that ALDH2 altered the redox status of cells by regulating acetaldehyde levels and that it further activated the AMP-activated protein kinase (AMPK) signaling pathway. CONCLUSION: Decreased levels of ALDH2 may indicate a poor prognosis in HCC patients, while forcing the expression of ALDH2 in HCC cells inhibited their aggressive behavior in vitro and in mice largely by modulating the activity of the ALDH2-acetaldehyde-redox-AMPK axis. Therefore, identifying ALDH2 expression levels in HCC might be a useful strategy for classifying HCC patients and for developing potential therapeutic strategies that specifically target metastatic HCC. (Hepatology 2017;65:1628-1644).


Subject(s)
Aldehyde Dehydrogenase, Mitochondrial/metabolism , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms, Experimental/enzymology , AMP-Activated Protein Kinases/metabolism , Animals , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , China/epidemiology , Ectopic Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Liver/pathology , Liver Neoplasms, Experimental/mortality , Liver Neoplasms, Experimental/pathology , Male , Mice, Nude , Middle Aged , Neoplasm Metastasis , Oxidation-Reduction , Random Allocation
7.
Cancer Lett ; 388: 1-11, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27894955

ABSTRACT

Cancer cells display altered metabolic phenotypes characterized by a high level of glycolysis, even under normoxic conditions. Because of a high rate of glycolytic flux and inadequate vascularization, tumor cells often suffer from nutrient deficiency and require metabolic adaptations to address such stresses. Although tumor-initiating cells (T-ICs) have been identified in various malignancies, the cells' metabolic phenotypes remain elusive. In this study, we observed that liver T-ICs preferentially survived under restricted glucose treatment. These cell populations compete successfully for glucose uptake by preferentially expressing glucose transporters (GLUT1 and GLUT3), whereas inhibition of GLUT1 or GLUT3 abolished the survival advantage and suppressed the tumorigenic potential of liver T-ICs. Among signaling pathways related to T-ICs, IL-6/STAT3 was identified to be responsible for the elevation of glucose uptake in liver T-ICs under glucose limitation. Further investigation revealed that IL-6 stimulation upregulated GLUT1 and GLUT3 expressions in CD133+ cells, particularly during glucose deprivation. More importantly, inhibition of glucose uptake sensitized liver T-ICs to sorafenib treatment and enhanced the therapeutic efficacy in vivo. Our findings suggest that blocking IL-6/STAT3-mediated preferential glucose uptake might be exploited for novel therapeutic targets during hepatocellular carcinoma (HCC) progression.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Antineoplastic Agents/administration & dosage , Carcinoma, Hepatocellular/pathology , Glucose/metabolism , Humans , Liver Neoplasms/pathology , Neoplastic Stem Cells/pathology , Niacinamide/administration & dosage , Niacinamide/therapeutic use , Phenylurea Compounds/administration & dosage , Sorafenib
9.
Oncoimmunology ; 5(9): e1183850, 2016.
Article in English | MEDLINE | ID: mdl-27757296

ABSTRACT

Despite their central function in tumor immunity, dendritic cells (DCs) can respond to inhibitory signals and become tolerogenic, curtailing T cell responses in vivo. Here, we provide the evidence for an inhibitory function of signal regulatory protein (SIRP) α in DC survival and activation. In tumors from human liver cancer patients, infiltrative DCs expressed elevated levels of SIRPα, which is correlated with the induction of immune tolerance within the tumors. Silencing of SIRPα resulted in a significant increase in the longevity of antigen-pulsed DCs in the draining lymph nodes. In addition, SIRPα controls the activation and output of DCs. Silencing of DC-expressed SIRPα induced spontaneous and enhanced production of IL12 and costimulatory molecules, resulting in more potent cytotoxic T lymphocyte responses, including the eradication of previously established solid tumors. SIRPα exerted such effects, at least in part, via the association and sequestration of p85 subunit of PI3K. Thus, SIRPα is a critical regulator of DC lifespan and activity, and its inhibition might improve the clinical efficacy of DC-based tumor vaccines.

10.
Exp Ther Med ; 12(4): 2554-2562, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27698758

ABSTRACT

Gut microbiota composition of patients with ulcerative colitis (UC) is markedly altered compared with healthy individuals. There is mounting evidence that probiotic therapy alleviates disease severity in animal models and patients with inflammatory bowel disease (IBD). Bacillus subtilisis, as a probiotic, has also demonstrated a protective effect in IBD. However, the therapeutic mechanism of its action has yet to be elucidated. In the present study, a dextrose sulfate sodium (DSS)-induced UC mouse model was used to investigate the role of B. subtilis in the restoration of gut flora and determine its effective dose. Mucosal damage was assessed by performing alcian blue staining, cytokine levels were analyzed by ELISA and microbiota composition was investigated using 454 pyrosequencing to target hypervariable regions V3-V4 of the bacterial 16S ribosomal RNA gene. The results demonstrated that a higher dose B. subtilisis administration ameliorated DSS-induced dysbiosis and gut inflammation by balancing beneficial and harmful bacteria and associated anti- and pro-inflammatory agents, thereby aiding intestinal mucosa recovery from DSS-induced injuries. These findings indicate that choosing the correct dose of B. subtilis is important for effective UC therapy. The present study also helped to elucidate the mechanisms of B. subtilis action and provided preclinical data for B. subtilis use in UC therapy.

11.
Nat Commun ; 7: 12992, 2016 10 05.
Article in English | MEDLINE | ID: mdl-27703150

ABSTRACT

Hepatitis B virus (HBV) can integrate into the human genome, contributing to genomic instability and hepatocarcinogenesis. Here by conducting high-throughput viral integration detection and RNA sequencing, we identify 4,225 HBV integration events in tumour and adjacent non-tumour samples from 426 patients with HCC. We show that HBV is prone to integrate into rare fragile sites and functional genomic regions including CpG islands. We observe a distinct pattern in the preferential sites of HBV integration between tumour and non-tumour tissues. HBV insertional sites are significantly enriched in the proximity of telomeres in tumours. Recurrent HBV target genes are identified with few that overlap. The overall HBV integration frequency is much higher in tumour genomes of males than in females, with a significant enrichment of integration into chromosome 17. Furthermore, a cirrhosis-dependent HBV integration pattern is observed, affecting distinct targeted genes. Our data suggest that HBV integration has a high potential to drive oncogenic transformation.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Hepatitis B virus/physiology , Liver Neoplasms/genetics , Liver Neoplasms/virology , Cell Transformation, Neoplastic , CpG Islands , DNA, Viral/genetics , Female , Genome, Human , Genome, Viral , Hepatitis B, Chronic/genetics , Humans , Kaplan-Meier Estimate , Liver Cirrhosis/genetics , Liver Cirrhosis/virology , Male , Real-Time Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Analysis, RNA , Virus Integration
12.
Oncotarget ; 7(6): 6711-26, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26744319

ABSTRACT

Due to a high rate of nutrient consumption and inadequate vascularization, hepatocellular carcinoma (HCC) cells constantly undergo metabolic stress during tumor development. Hepatitis B virus (HBV) X protein (HBx) has been implicated in the pathogenesis of HBV-induced HCC. In this study, we investigated the functional roles of HBx in HCC adaptation to metabolic stress. Up-regulation of HBx increased the intracellular ATP and NADPH generation, and induced the resistance to glucose deprivation, whereas depletion of HBx via siRNA abolished these effects and conferred HCC cells sensitive to glucose restriction. Though HBx did not affect the glycolysis and oxidative phosphorylation capacity of HCC cells under normal culture conditions, it facilitated fatty acid oxidation (FAO) in the absence of glucose, which maintained NADPH and ATP levels. Further investigation showed that HBx expression, under glucose deprivation, stimulated phosphorylation of AMP-activated protein kinase (AMPK) and acetyl-CoA carboxylase (ACC) via a calcium/CaMKK-dependent pathway, which was required for the activation of FAO. Conversely, inhibition of FAO by etomoxir (ETO) restored the sensitivity of HBx-expressing cells to glucose deficiency in vitro and retarded xenograft tumor formation in vivo. Finally, HBx-induced activation of the AMPK and FAO pathways were also observed in xenograft tumors and HBV-associated HCC specimens. Our data suggest that HBx plays a key role in the maintenance of redox and energy homeostasis by activating FAO, which is critical for HCC cell survival under conditions of metabolic stress and might be exploited for therapeutic benefit.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Fatty Acids/metabolism , Liver Neoplasms/metabolism , Trans-Activators/metabolism , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/physiology , Glucose/deficiency , Glucose/metabolism , Glycolysis , Hep G2 Cells , Heterografts , Humans , Lipid Metabolism , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , NADP/metabolism , Oxidation-Reduction , Oxidative Phosphorylation , Stress, Physiological , Trans-Activators/biosynthesis , Trans-Activators/genetics , Up-Regulation , Viral Regulatory and Accessory Proteins
13.
Hepatology ; 63(4): 1272-86, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26698170

ABSTRACT

UNLABELLED: Solid tumors often suffer from suboptimal oxygen and nutrient supplies. This stress underlies the requirement for metabolic adaptation. Aberrantly activated de novo lipogenesis is critical for development and progression of human hepatocellular carcinoma (HCC). However, whether de novo lipogenesis influences biological behaviors of HCCs under conditions of metabolic stress are still poorly understood. Here, we show that HCCs display distinct levels of glucose-derived de novo lipogenesis, which are positively correlated with their survival responses to glucose limitation. The enhanced lipogenesis in HCCs is characterized by an increased expression of rate-limiting enzyme acetyl-coenzyme A carboxylase alpha (ACCα). ACCα-mediated fatty acid (FA) synthesis determines the intracellular lipid content that is required to maintain energy hemostasis and inhibit cell death by means of FA oxidation (FAO) during metabolic stress. In accord, overexpression of ACCα facilitates tumor growth. ACCα forms a complex with carnitine palmitoyltransferase 1A (CPT1A) and prevents its mitochondria distribution under nutrient-sufficient conditions. During metabolic stress, phosphorylation of ACCα leads to dissociation of the complex and mitochondria localization of CPT1A, thus promoting FAO-mediated cell survival. Therefore, ACCα could provide both the substrate and enzyme storage for FAO during glucose deficiency. Up-regulation of ACCα is also significantly correlated with poorer overall survival and disease recurrence postsurgery. Multivariate Cox's regression analysis identified ACCα as an effective predictor of poor prognosis. CONCLUSION: These results present novel mechanistic insight into a pivotal role of ACCα in maintaining HCC survival under metabolic stress. It could be exploited as a novel diagnostic marker and therapeutic target.


Subject(s)
Acetyl-CoA Carboxylase/metabolism , Carcinoma, Hepatocellular/enzymology , Glucose/metabolism , Liver Neoplasms/enzymology , Oxidative Stress , Acetyl-CoA Carboxylase/genetics , Animals , Apoptosis/genetics , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Survival/genetics , Disease Models, Animal , Fatty Acid Synthases/metabolism , Fatty Acids/metabolism , Gene Expression Regulation, Neoplastic , Gene Silencing , Heterografts , Humans , Kaplan-Meier Estimate , Lipid Metabolism/physiology , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , Predictive Value of Tests , Prognosis , Proportional Hazards Models , Survival Analysis , Up-Regulation
14.
Nat Commun ; 6: 8457, 2015 Oct 07.
Article in English | MEDLINE | ID: mdl-26443326

ABSTRACT

Merlin, which is encoded by the tumour suppressor gene Nf2, plays a crucial role in tumorigenesis and metastasis. However, little is known about the functional importance of Merlin splicing forms. In this study, we show that Merlin is present at low levels in human hepatocellular carcinoma (HCC), particularly in metastatic tumours, where it is associated with a poor prognosis. Surprisingly, a splicing variant of Merlin that lacks exons 2, 3 and 4 ((Δ2-4)Merlin) is amplified in HCC and portal vein tumour thrombus (PVTT) specimens and in the CSQT2 cell line derived from PVTT. Our studies show that (Δ2-4)Merlin interferes with the capacity of wild-type Merlin to bind ß-catenin and ERM, and it is expressed in the cytoplasm rather than at the cell surface. Furthermore, (Δ2-4)Merlin overexpression increases the expression levels of ß-catenin and stemness-related genes, induces the epithelium-mesenchymal-transition phenotype promoting cell migration in vitro and the formation of lung metastasis in vivo. Our results indicate that the (Δ2-4)Merlin variant disrupts the normal function of Merlin and promotes tumour metastasis.


Subject(s)
Alternative Splicing , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , Lung Neoplasms/genetics , Neurofibromin 2/genetics , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Cell Line, Tumor , Cytoplasm/metabolism , Cytoskeletal Proteins/metabolism , Epithelial-Mesenchymal Transition/genetics , Female , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Male , Membrane Proteins/metabolism , Microfilament Proteins/metabolism , Middle Aged , Neoplasm Metastasis , Neurofibromin 2/metabolism , Portal Vein/pathology , Thrombosis/pathology , beta Catenin/metabolism
15.
Clin Res Hepatol Gastroenterol ; 38(3): 337-45, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24502885

ABSTRACT

BACKGROUND/AIMS: A major hepatectomy occasionally lead to acute liver failure and death. We demonstrated the anti-oxidative and anti-inflammatory effects and functional mechanisms of hydrogen-rich saline (HS), a novel antioxidant, on an experimental model of rats after a partial hepatectomy (PH). METHODS: The rats underwent a 90% hepatectomy. HS was given intraperitoneally after the operation and every 8hours after. RESULTS: HS markedly improved the survival rate of two experimental groups after the massive hepatectomy and inhibited increases in serum levels of TBIL, DBIL, ALT and AST. The histopathological analysis demonstrated that HS attenuated inflammatory changes in the liver. HS administration markedly lowered the massive hepatectomy induced elevation of the serum hyaluronic acid (HA) concentrations. HS inhibited the formation of one of the markers of oxidative damage, malondialdehyde (MDA), and increased the activities of superoxide dismutase (SOD) in liver tissue. In the HS-treated group, increases in inflammatory cytokines, such as TNF-α, IL-6 and HMGB-1, were inhibited in the liver tissue. The NF-κB p65 staining revealed that HS inhibited the activation of the transcription factor nuclear factor kappa B (NF-kB). CONCLUSIONS: HS attenuates the massive hepatectomy induced liver injury not only by attenuating oxidative damage, but also by reducing the production of inflammatory cytokines, such as TNF-α, IL-6 and HMGB-1, in part through the inhibition of NF-kB activation.


Subject(s)
Hepatectomy/adverse effects , Hydrogen/pharmacology , Liver Failure/prevention & control , Sodium Chloride/pharmacology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Bilirubin/blood , Cytokines/metabolism , Hyaluronic Acid/blood , Liver/metabolism , Malondialdehyde/metabolism , Models, Animal , NF-kappa B/metabolism , Rats, Sprague-Dawley , Superoxide Dismutase/metabolism
16.
J Hepatol ; 59(4): 762-8, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23714159

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) develops in response to chronic hepatic injury. Although induced cell death is regarded as the major component of p53 tumor-suppressive activity, we recently found that sustained p53 activation subsequent to DNA damage promotes inflammation-associated hepatocarcinogenesis. Here we aim at exploring the mechanism linking p53 activation and hepatic inflammation during hepatocarcinogenesis. METHODS: p53(-/-) hepatocytes expressing inducible p53 and primary wild type hepatocytes were treated to induce p53 expression. The supernatants were collected and analyzed for the presence of released inflammatory cytokines. Ethyl pyruvate was used in a rat model of carcinogen-induced hepatocarcinogenesis to examine its effect on p53-dependent chronic hepatic injury, inflammation, and tumorigenesis. RESULTS: Here we show that cytoplasmic translocation and circulating levels of potent inflammatory molecule high-mobility group protein 1 (HMGB1) were greater in wild type rats than in p53(+/-) rats following carcinogen administration. Restoration of p53 expression in p53-null hepatocytes or induction of endogenous p53 in wild type hepatocytes gives rise to the release of HMGB1. Administration of the HMGB1 release inhibitor ethyl pyruvate, which does not affect p53-mediated hepatic apoptosis, substantially prevented carcinogen-induced cirrhosis and tumorigenesis in rat livers. CONCLUSIONS: These results suggest that although p53 is usually regarded as a tumor suppressor, its constant activation can promote pro-tumorigenic inflammation, at least in part, via inducing HMGB1 release. Application of HMGB1 inhibitors when restoring p53 in cancer therapy might protect against pro-tumorigenic effects while leaving p53-mediated clearance of malignant cells intact.


Subject(s)
Genes, p53 , HMGB1 Protein/metabolism , Liver Neoplasms, Experimental/etiology , Animals , Cell Line , Diethylnitrosamine/toxicity , Gene Knockout Techniques , Hepatitis, Chronic/etiology , Hepatitis, Chronic/metabolism , Hepatitis, Chronic/pathology , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Rats , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism
17.
J Hepatol ; 57(4): 803-12, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22727732

ABSTRACT

BACKGROUND & AIMS: Due to its anatomic connection, the liver is constantly exposed to gut-derived bacterial products or metabolites. Disruption of gut homeostasis is associated with many human diseases. The aim of this study was to determine the role of gut homeostasis in initiation and progression of hepatocellular carcinoma (HCC). METHODS: Disruption of intestinal homeostasis by penicillin or dextran sulfate sodium (DSS) and its restoration by probiotics were applied in a diethylnitrosamine (DEN) model of rat hepatocarcinogenesis. RESULTS: Patients with liver cirrhosis and HCC had significantly increased serum endotoxin levels. Chronic DEN treatment of rats was associated with an imbalance of subpopulations of the gut microflora including a significant suppression of Lactobacillus species, Bifidobacterium species and Enterococcus species as well as intestinal inflammation. Induction of enteric dysbacteriosis or intestinal inflammation by penicillin or DSS, respectively, significantly promoted tumor formation. Administration of probiotics dramatically mitigated enteric dysbacteriosis, ameliorated intestinal inflammation, and most importantly, decreased liver tumor growth and multiplicity. Interestingly, probiotics not only inhibited the translocation of endotoxin, which bears pathogen-associated molecular patterns (PAMPs) but also the activation of damage-associated molecular patterns (DAMPs) such as high-mobility group box 1 (HMGB1). As a result, the production of pro- and anti-inflammatory cytokines was skewed in favor of a reduced tumorigenic inflammation in the liver. CONCLUSIONS: The data highlights the importance of gut homeostasis in the pathogenesis of HCC. Modulation of the gut microbiota by probiotics may represent a new avenue for therapeutic intervention to treat or prevent HCC development.


Subject(s)
Carcinoma, Hepatocellular/pathology , Endotoxins/metabolism , Gastrointestinal Tract/microbiology , Homeostasis , Liver Neoplasms, Experimental/pathology , Probiotics/pharmacology , Alkylating Agents/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Bifidobacterium/drug effects , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/etiology , Cytokines/biosynthesis , Dextran Sulfate/pharmacology , Diethylnitrosamine/pharmacology , Diethylnitrosamine/toxicity , Disease Progression , Endotoxins/blood , Enterococcus/drug effects , Gastroenteritis/chemically induced , Gastroenteritis/drug therapy , Gastroenteritis/metabolism , Gastrointestinal Tract/physiopathology , HMGB1 Protein/metabolism , Homeostasis/drug effects , Humans , Lactobacillus/drug effects , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/microbiology , Male , Penicillins/pharmacology , Probiotics/therapeutic use , Rats , Rats, Sprague-Dawley
18.
J Hepatol ; 57(3): 613-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22612999

ABSTRACT

BACKGROUND & AIMS: Accumulating evidence suggests the involvement of tumor-initiating cells (T-ICs) in cancer genesis, but whether liver T-ICs contribute to HCC invasion and metastasis remains unclear. METHODS: OV6(+) T-ICs were isolated from SMMC7721 and HuH7 cell lines by magnetic sorting. Characteristics of T-ICs were assessed by in vitro and mouse xenograft assays. Expression of OV6 was determined by immunostaining in specimens from 218 HCC patients, and Kaplan-Meier survival analysis was used to determine the correlation of OV6 expression with prognosis. RESULTS: OV6(+) T-ICs isolated from HCC cell lines not only possess a higher capacity to form tumor spheroids in vitro, but also had a greater potential to form tumors when implanted in non-obese diabetic/severe combined immunodeficient mice, suggesting their elevated self-renewal capacity and tumorigenicity. Moreover, OV6(+) T-ICs exhibited more invasive and metastatic potentials both in vitro and in vivo. Patients with more OV6(+) tumor cells were associated with aggressive clinicopathologic features and poor prognosis. CXCR4 is expressed at higher levels in OV6(+) cells. Recombinant stromal cell-derived factor-1 (SDF-1) treatment expanded the OV6(+) HCC T-ICs population, by sustaining the stem cell property of OV6(+) cells. The SDF-1 effect was blocked by a specific CXCR4 inhibitor, AMD3100, or transfection of siRNA targeting CXCR4. CONCLUSIONS: OV6(+) HCC cells may represent a subpopulation of T-ICs with augmented invasion and metastasis potential, which contribute to progression and metastasis of HCC. The SDF-1/CXCR4 axis also provides therapeutic targets for elimination of liver T-ICs.


Subject(s)
Antigens, Differentiation/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , AC133 Antigen , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Benzylamines , Cell Adhesion Molecules/metabolism , Cell Line, Tumor , Cell Movement , Chemokine CXCL12/metabolism , Cyclams , Disease Progression , Disease-Free Survival , Epithelial Cell Adhesion Molecule , Female , Gene Knockdown Techniques , Glycoproteins/metabolism , Heterocyclic Compounds/pharmacology , Humans , Kaplan-Meier Estimate , Lung Neoplasms/secondary , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Peptides/metabolism , Prognosis , RNA, Small Interfering , Receptors, CXCR4/drug effects , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Spheroids, Cellular
19.
Cancer Prev Res (Phila) ; 5(9): 1090-102, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22617167

ABSTRACT

Robust clinical and epidemiologic data support the role of inflammation as a key player in hepatocellular carcinoma (HCC) development. Our previous data showed that gut-derived lipopolysaccharide (LPS) promote HCC development by activating Toll-like receptor 4 (TLR4) expressed on myeloid-derived cells. However, the effects of gut-derived LPS on other types of liver injury models are yet to be studied. The purpose of this study was to determine the importance of gut-derived LPS and TLR4 signaling in a T-cell-mediated hepatitis-Con A-induced hepatitis model, which mimic the viral hepatitis. Reduction of endotoxin using antibiotics regimen or genetic ablation of TLR4 in mice significantly alleviate Con A-induced liver injury by inhibiting the infiltration of T lymphocytes into the liver and the activation of CD4(+) T lymphocytes as well as the production of T helper 1 cytokines; in contrast, exogenous LPS can promote Con A-induced hepatitis and CD4(+) T cells activation in vivo and in vitro. Reconstitution of TLR4-expressing myeloid cells in TLR4-deficient mice restored Con A-induced liver injury and inflammation, indicating the major cell target of LPS. In addition, TLR4 may positively regulate the target hepatocellular apoptosis via the perforin/granzyme B pathway. These data suggest that gut-derived LPS and TLR4 play important positive roles in Con A-induced hepatitis and modulation of the gut microbiotia may represent a new avenue for therapeutic intervention to treat acute hepatitis induced by hepatitis virus infection, thus to prevent hepatocellular carcinoma.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hepatitis/immunology , Lipopolysaccharides/immunology , Lymphocyte Activation/immunology , Toll-Like Receptor 4/immunology , Animals , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/metabolism , Disease Models, Animal , Flow Cytometry , Intestines/immunology , Intestines/microbiology , Liver Neoplasms/immunology , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Signal Transduction/immunology , Toll-Like Receptor 4/metabolism
20.
Hepatology ; 55(1): 108-20, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21932402

ABSTRACT

UNLABELLED: Hepatitis B virus X (HBx) protein is implicated in hepatitis B virus (HBV)-associated liver carcinogenesis. However, it remains unclear whether HBx-expressing hepatic progenitor cells (HPCs) are attributed to liver tumor formation. In this study, by using HBx transgenic mice and a 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced liver injury model, the relationship between HBx expression and tumorigenicity of HPCs was analyzed. Compared with control mice, an elevated number of EpCAM(+) cells with characteristics of HPCs was observed in HBx mice after 1 month and 4 months of DDC diet feeding. All HBx transgenic mice developed liver tumors characterized by histological features of both hepatocellular carcinoma (HCC) and cholangiocarcinoma after 7 months of DDC feeding. Notably, EpCAM(+) HPCs isolated from premalignant HBx mice exposed to a DDC diet for 4 months formed subcutaneous mixed-lineage tumors (four out of six) in nonobese diabetic/severe-combined immunodeficient (NOD/SCID) mice, and none of the cells from wildtype (WT) induced tumor, indicating that HBx may induce malignant transformation of HPCs that contributes to tumorigenesis. We also found higher titers of circulating interleukin (IL)-6, activities of IL-6/STAT3, and Wnt/ß-catenin signaling pathways in HBx transgenic mice, suggesting HBx may induce intrinsic changes in HPCs by way of the above signaling that enables HPCs with tumorigenicity potential. Finally, clinical evidence showed that high HBx expression in human HBV-related HCC was statistically associated with expansion of EpCAM(+) or OV6(+) tumor cells and aggressive clinicopathologic features. CONCLUSION: HBx induces intrinsic cellular transformation promoting the expansion and tumorigenicity of HPCs in DDC-treated mice, which may be a possible origin for liver cancer induced by chronic hepatitis infection.


Subject(s)
Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/virology , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/virology , Pyridines/toxicity , Trans-Activators/genetics , Animals , Bile Duct Neoplasms/chemically induced , Bile Duct Neoplasms/physiopathology , Bile Duct Neoplasms/virology , Bile Ducts, Intrahepatic , Carcinoma, Hepatocellular/physiopathology , Cell Transformation, Neoplastic/chemically induced , Cholangiocarcinoma/chemically induced , Cholangiocarcinoma/physiopathology , Cholangiocarcinoma/virology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Humans , Liver Neoplasms, Experimental/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Middle Aged , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cells/drug effects , Stem Cells/physiology , Stem Cells/virology , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...