Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cancer Metastasis Rev ; 42(3): 677-698, 2023 09.
Article in English | MEDLINE | ID: mdl-37432606

ABSTRACT

Cancer is a multi-step process that can be viewed as a cellular and immunological shift away from homeostasis in response to selected infectious agents, mutations, diet, and environmental carcinogens. Homeostasis, which contributes importantly to the definition of "health," is maintained, in part by the production of short-chain fatty acids (SCFAs), which are metabolites of specific gut bacteria. Alteration in the composition of gut bacteria, or dysbiosis, is often a major risk factor for some two dozen tumor types. Dysbiosis is often characterized by diminished levels of SCFAs in the stool, and the presence of a "leaky gut," permitting the penetration of microbes and microbial derived molecules (e.g., lipopolysaccharides) through the gut wall, thereby triggering chronic inflammation. SCFAs attenuate inflammation by inhibiting the activation of nuclear factor kappa B, by decreasing the expression of pro-inflammatory cytokines such as tumor necrosis factor alpha, by stimulating the expression of anti-inflammatory cytokines such as interleukin-10 and transforming growth factor beta, and by promoting the differentiation of naĆÆve T cells into T regulatory cells, which down-regulate immune responses by immunomodulation. SCFA function epigenetically by inhibiting selected histone acetyltransferases that alter the expression of multiple genes and the activity of many signaling pathways (e.g., Wnt, Hedgehog, Hippo, and Notch) that contribute to the pathogenesis of cancer. SCFAs block cancer stem cell proliferation, thereby potentially delaying or inhibiting cancer development or relapse by targeting genes and pathways that are mutated in tumors (e.g., epidermal growth factor receptor, hepatocyte growth factor, and MET) and by promoting the expression of tumor suppressors (e.g., by up-regulating PTEN and p53). When administered properly, SCFAs have many advantages compared to probiotic bacteria and fecal transplants. In carcinogenesis, SCFAs are toxic against tumor cells but not to surrounding tissue due to differences in their metabolic fate. Multiple hallmarks of cancer are also targets of SCFAs. These data suggest that SCFAs may re-establish homeostasis without overt toxicity and either delay or prevent the development of various tumor types.


Subject(s)
Dysbiosis , Neoplasms , Humans , Cytokines/metabolism , Inflammation/metabolism , Neoplasms/etiology , Fatty Acids, Volatile/metabolism
2.
Semin Cancer Biol ; 35 Suppl: S25-S54, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25892662

ABSTRACT

Proliferation is an important part of cancer development and progression. This is manifest by altered expression and/or activity of cell cycle related proteins. Constitutive activation of many signal transduction pathways also stimulates cell growth. Early steps in tumor development are associated with a fibrogenic response and the development of a hypoxic environment which favors the survival and proliferation of cancer stem cells. Part of the survival strategy of cancer stem cells may manifested by alterations in cell metabolism. Once tumors appear, growth and metastasis may be supported by overproduction of appropriate hormones (in hormonally dependent cancers), by promoting angiogenesis, by undergoing epithelial to mesenchymal transition, by triggering autophagy, and by taking cues from surrounding stromal cells. A number of natural compounds (e.g., curcumin, resveratrol, indole-3-carbinol, brassinin, sulforaphane, epigallocatechin-3-gallate, genistein, ellagitannins, lycopene and quercetin) have been found to inhibit one or more pathways that contribute to proliferation (e.g., hypoxia inducible factor 1, nuclear factor kappa B, phosphoinositide 3 kinase/Akt, insulin-like growth factor receptor 1, Wnt, cell cycle associated proteins, as well as androgen and estrogen receptor signaling). These data, in combination with bioinformatics analyses, will be very important for identifying signaling pathways and molecular targets that may provide early diagnostic markers and/or critical targets for the development of new drugs or drug combinations that block tumor formation and progression.


Subject(s)
Cell Cycle Proteins/genetics , Cell Proliferation/drug effects , Neoplasms/pathology , Neoplasms/therapy , Antineoplastic Agents/therapeutic use , Cell Cycle Proteins/biosynthesis , Epithelial-Mesenchymal Transition/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Signal Transduction/drug effects
3.
Semin Cancer Biol ; 35 Suppl: S276-S304, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26590477

ABSTRACT

Targeted therapies and the consequent adoption of "personalized" oncology have achieved notable successes in some cancers; however, significant problems remain with this approach. Many targeted therapies are highly toxic, costs are extremely high, and most patients experience relapse after a few disease-free months. Relapses arise from genetic heterogeneity in tumors, which harbor therapy-resistant immortalized cells that have adopted alternate and compensatory pathways (i.e., pathways that are not reliant upon the same mechanisms as those which have been targeted). To address these limitations, an international task force of 180 scientists was assembled to explore the concept of a low-toxicity "broad-spectrum" therapeutic approach that could simultaneously target many key pathways and mechanisms. Using cancer hallmark phenotypes and the tumor microenvironment to account for the various aspects of relevant cancer biology, interdisciplinary teams reviewed each hallmark area and nominated a wide range of high-priority targets (74 in total) that could be modified to improve patient outcomes. For these targets, corresponding low-toxicity therapeutic approaches were then suggested, many of which were phytochemicals. Proposed actions on each target and all of the approaches were further reviewed for known effects on other hallmark areas and the tumor microenvironment. Potential contrary or procarcinogenic effects were found for 3.9% of the relationships between targets and hallmarks, and mixed evidence of complementary and contrary relationships was found for 7.1%. Approximately 67% of the relationships revealed potentially complementary effects, and the remainder had no known relationship. Among the approaches, 1.1% had contrary, 2.8% had mixed and 62.1% had complementary relationships. These results suggest that a broad-spectrum approach should be feasible from a safety standpoint. This novel approach has potential to be relatively inexpensive, it should help us address stages and types of cancer that lack conventional treatment, and it may reduce relapse risks. A proposed agenda for future research is offered.


Subject(s)
Genetic Heterogeneity , Molecular Targeted Therapy , Neoplasms/therapy , Precision Medicine , Antineoplastic Agents, Phytogenic/therapeutic use , Drug Resistance, Neoplasm/genetics , Humans , Neoplasms/genetics , Neoplasms/pathology , Neoplasms/prevention & control , Signal Transduction , Tumor Microenvironment/genetics
4.
Article in English | MEDLINE | ID: mdl-38317750

ABSTRACT

Background: Transmembrane protein 43 (TMEM43), a member of the TMEM subfamily, is encoded by a highly conserved gene and widely expressed in most species from bacteria to humans. In previous studies, TMEM43 has been found to play an important role in a variety of tumors. However, the role of TMEM43 in cancer remains unclear. Methods: We utilized the RNA sequencing (RNA-seq) and The Cancer Genome Atlas (TGCA) databases to explore and identify genes that may play an important role in the occurrence and development of hepatocellular carcinoma (HCC), such as TMEM43. The role of TMEM43 in HCC was explored through Cell Counting Kit-8 (CCK-8) cloning, flow cytometry, and Transwell experiments. The regulatory relationship between TMEM43 and voltage-dependent anion channel 1 (VDAC1) was investigated through coimmunoprecipitation (co-IP) and western blot (WB) experiments. WB was used to study the deubiquitination effect of ubiquitin-specific protease 7 (USP7) on TMEM43. Results: In this study, we utilized the RNA-seq and TGCA databases to mine data and found that TMEM43 is highly expressed in HCC. The absence of TMEM43 in cancer cells was shown to inhibit tumor development. Further research detected an important regulatory relationship between TMEM43 and VDAC1. In addition, we found that USP7 affected the progression of HCC by regulating the ubiquitination level of TMEM43 through deubiquitination. Conclusions: Our study demonstrated that USP7 participates in the growth of HCC tumors through TMEM43/VDAC1.Our results suggest that USP7/TMEM43/VDAC1 may have predictive value and represent a new treatment strategy for HCC.

5.
Antimicrob Agents Chemother ; 57(11): 5344-54, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23959305

ABSTRACT

Hepatitis B virus (HBV)-associated chronic liver diseases are treated with nucleoside analogs that target the virus polymerase. While these analogs are potent, drugs are needed to target other virus-encoded gene products to better block the virus replication cycle and chronic liver disease. This work further characterized GLS4 and compared it to the related BAY 41-4109, both of which trigger aberrant HBV core particle assembly, where the virus replication cycle occurs. This was done in HepAD38 cells, which replicate HBV to high levels. In vitro, GLS4 was significantly less toxic for primary human hepatocytes (P < 0.01 up to 100 ĀµM), inhibited virus accumulation in the supernantant of HepAD38 cells (P < 0.02 up to 100 nM), inhibited HBV replicative forms in the liver with a significantly lower 50% effective concentration (EC50) (P < 0.02), and more strongly inhibited core gene expression (P < 0.001 at 100 to 200 nM) compared to BAY 41-4109. In vivo characterization was performed in nude mice inoculated with HepAD38 cells, which grew out as tumors, resulting in viremia. Treatment of mice with GLS4 and BAY 41-4109 showed strong and sustained suppression of virus DNA to about the same extents both during and after treatment. Both drugs reduced the levels of intracellular core antigen in the tumors. Alanine aminotransferase levels were normal. Tumor and total body weights were not affected by treatment. Thus, GLS4 was as potent as the prototype, BAY 41-4109, and was superior to lamivudine, in that there was little virus relapse after the end of treatment and no indication of toxicity.


Subject(s)
Antiviral Agents/pharmacology , DNA, Viral/antagonists & inhibitors , Hepatitis B virus/drug effects , Hepatitis B, Chronic/drug therapy , Pyrimidines/pharmacology , Thiazoles/pharmacology , Viremia/drug therapy , Virion/drug effects , Animals , Antiviral Agents/chemical synthesis , Cell Line , DNA, Viral/biosynthesis , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Hepatitis B virus/growth & development , Hepatitis B virus/ultrastructure , Hepatitis B, Chronic/virology , Hepatocytes/drug effects , Hepatocytes/pathology , Hepatocytes/virology , Humans , Lamivudine/pharmacology , Male , Mice , Mice, Nude , Microbial Sensitivity Tests , Primary Cell Culture , Pyridines/pharmacology , Pyrimidines/chemical synthesis , Thiazoles/chemical synthesis , Viral Core Proteins/antagonists & inhibitors , Viral Core Proteins/metabolism , Viremia/virology , Virion/ultrastructure , Virus Replication/drug effects
6.
BMC Cancer ; 13: 167, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23537372

ABSTRACT

BACKGROUND: UCN-01 (7-hydroxystaurosporine), a protein kinase inhibitor, has attracted a great deal of attention as a potent antitumour agent. Several clinical trials of UCN-01 alone or in combination with other agents for different tumour types are currently underway, and some of these trials have had positive results. Hepatocellular carcinoma has high incidence rates and is associated with poor prognosis and high mortality rates. METHODS: Three different hepatoma cell lines (Huh7, HepG2, and Hep3B) were treated with different concentrations of UCN-01, and the anti-tumour effects of UCN-01 were evaluated. Following UCN-01 treatment, cell growth was measured using an MTT assay, cell cycle arrest was assayed using flow cytometry, and the mechanisms of cell cycle arrest and invasion inhibition were investigated through western blotting and a Matrigel invasion assay. RESULTS: After a 72-h UCN-01 treatment, the growth of different hepatoma cell lines was significantly inhibited in a dose-dependent manner, with IC50 values ranging from 69.76 to 222.74 nM. Flow cytometry results suggested that UCN-01 inhibits proliferation in the hepatoma cells by inducing S and G2/M phase arrest, but not G1/S arrest, which differs from previous reports that used other tumour cell lines. Western blot results illustrated that UCN-01 induces a G2/M phase arrest, regardless of the status of the p53/P21(waf1) pathway, whereas the CHK2/CDC25C pathway and the p53/p21(waf1)pathway were involved in the UCN-01-induced S phase arrest. UCN-01 remarkably inhibited Huh7 cell invasion in a time-dependent manner. Suppression of Huh7 cell invasion may be due to the down-regulation of phosphorylated Ɵ-catenin by UCN-01. CONCLUSIONS: These findings suggest that UCN-01 induces hepatoma cell growth inhibition by regulating the p53/p21(waf1) and CHK2/CDC25 pathways. Suppression of Huh7 cell invasion by UCN-01 may be due to the down-regulation of phosphorylated Ɵ-catenin. These data lend support for further studies on UCN-01 as a promising anti-HCC candidate.


Subject(s)
Cell Cycle Checkpoints/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Staurosporine/analogs & derivatives , Tumor Suppressor Protein p53/metabolism , cdc25 Phosphatases/metabolism , Antineoplastic Agents/pharmacology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Checkpoint Kinase 2 , Humans , Inhibitory Concentration 50 , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Staurosporine/pharmacology , Tumor Suppressor Protein p53/genetics
7.
Antimicrob Agents Chemother ; 56(12): 6186-91, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22985879

ABSTRACT

Next-generation therapies for chronic hepatitis B virus (HBV) infection will involve combinations of established and/or experimental drugs. The current study investigated the in vitro and in vivo efficacy of tenofovir disoproxil fumarate (TDF) and/or emtricitabine [(-)-FTC] alone and in combination therapy for HBV infection utilizing the HepAD38 system (human hepatoblastoma cells transfected with HBV). Cellular pharmacology studies demonstrated increased levels of (-)-FTC triphosphate with coincubation of increasing concentrations of TDF, while (-)-FTC had no effect on intracellular tenofovir (TFV) diphosphate levels. Quantification of extracellular HBV by real-time PCR from hepatocytes demonstrated the anti-HBV activity with TDF, (-)-FTC, and their combination. Combination of (-)-FTC with TDF or TFV (ratio, 1:1) had a weighted average combination index of 0.7 for both combination sets, indicating synergistic antiviral effects. No cytotoxic effects were observed with any regimens. Using an in vivo murine model which develops robust HBV viremia in nude mice subcutaneously injected with HepAD38 cells, TDF (33 to 300 mg/kg of body weight/day) suppressed virus replication for up to 10 days posttreatment. At 300 mg/kg/day, (-)-FTC strongly suppressed virus titers to up to 14 days posttreatment. Combination therapy (33 mg/kg/day each drug) sustained suppression of virus titer/ml serum (<1 log(10) unit from pretreatment levels) at 14 days posttreatment, while single-drug treatments yielded virus titers 1.5 to 2 log units above the initial virus titers. There was no difference in mean alanine aminotransferase values or mean wet tumor weights for any of the groups, suggesting a lack of drug toxicity. TDF-(-)-FTC combination therapy provides more effective HBV suppression than therapy with each drug alone.


Subject(s)
Adenine/analogs & derivatives , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Deoxycytidine/analogs & derivatives , Hepatitis B virus/drug effects , Hepatitis B/drug therapy , Organophosphonates/pharmacology , Organophosphonates/therapeutic use , Reverse Transcriptase Inhibitors/pharmacology , Virus Replication/drug effects , Adenine/pharmacology , Adenine/therapeutic use , Adenine/toxicity , Animals , Antiviral Agents/toxicity , Cell Line , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Deoxycytidine/toxicity , Drug Combinations , Drug Interactions , Emtricitabine , Hepatitis B/virology , Mice , Mice, Nude , Organophosphonates/toxicity , Phosphorylation , Polymerase Chain Reaction , Reverse Transcriptase Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/toxicity , Tenofovir , Viremia/drug therapy , Viremia/virology
8.
J Virol ; 85(6): 2793-802, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21177818

ABSTRACT

Hepatitis C virus (HCV)-related research has been hampered by the lack of appropriate small-animal models. It has been reported that tree shrews, or tupaias (Tupaia belangeri), can be infected with serum-derived HCV. However, these reports do not firmly establish the tupaia as a reliable model of HCV infection. Human CD81, scavenger receptor class B type I (SR-BI), claudin 1 (CLDN1), and occludin (OCLN) are considered essential receptors or coreceptors for HCV cell entry. In the present study, the roles of these tupaia orthologs in HCV infection were assessed. Both CD81 and SR-BI of tupaia were found to be able to bind with HCV envelope protein 2 (E2). In comparison with human CD81, tupaia CD81 exhibited stronger binding activity with E2 and increased HCV pseudoparticle (HCVpp) cell entry 2-fold. The 293T cells transfected with tupaia CLDN1 became susceptible to HCVpp infection. Moreover, simultaneous transfection of the four tupaia factors into mouse NIH 3T3 cells made the cells susceptible to HCVpp infection. HCVpp of diverse genotypes were able to infect primary tupaia hepatocytes (PTHs), and this infection could be blocked by either anti-CD81 or anti-SR-BI. PTHs could be infected by cell culture-produced HCV (HCVcc) and did produce infectious progeny virus in culture supernatant. These findings indicate that PTHs possess all of the essential factors required for HCV entry and support the complete HCV infection cycle. This highlights both the mechanisms of susceptibility of tupaia to HCV infection and the possibility of using tupaia as a promising small-animal model in HCV study.


Subject(s)
Antigens, CD/metabolism , CD36 Antigens/metabolism , Hepacivirus/physiology , Membrane Proteins/metabolism , Tupaia/virology , Viral Envelope Proteins/metabolism , Animals , Cell Line , Claudin-1 , Disease Models, Animal , Humans , Mice , Molecular Sequence Data , Occludin , Protein Binding , Sequence Analysis, DNA , Tetraspanin 28 , Virus Internalization
9.
Biomedicines ; 10(9)2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36140311

ABSTRACT

Patients who are carriers of the hepatitis B virus (HBV) are at high risk of chronic liver disease (CLD) which proceeds from hepatitis, to fibrosis, cirrhosis and to hepatocellular carcinoma (HCC). The hepatitis B-encoded X antigen, HBx, promotes virus gene expression and replication, protects infected hepatocytes from immunological destruction, and promotes the development of CLD and HCC. For virus replication, HBx regulates covalently closed circular (ccc) HBV DNA transcription, while for CLD, HBx triggers cellular oxidative stress, in part, by triggering mitochondrial damage that stimulates innate immunity. Constitutive activation of NF-κB by HBx transcriptionally activates pro-inflammatory genes, resulting in hepatocellular destruction, regeneration, and increased integration of the HBx gene into the host genome. NF-κB is also hepatoprotective, which sustains the survival of infected cells. Multiple therapeutic approaches include direct-acting anti-viral compounds and immune-stimulating drugs, but functional cures were not achieved, in part, because none were yet devised to target HBx. In addition, many patients with cirrhosis or HCC have little or no virus replication, but continue to express HBx from integrated templates, suggesting that HBx contributes to the pathogenesis of CLD. Blocking HBx activity will, therefore, impact multiple aspects of the host-virus relationship that are relevant to achieving a functional cure.

10.
Int J Cancer ; 128(12): 2980-93, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-20725996

ABSTRACT

Hepatocellular carcinoma (HCC) is the second most common malignancy in Asia, with a 5-year survival rate of less than 5% due to high recurrence after surgery and resistance to chemotherapy. A variety of therapeutic interventions to treat HCC, particularly gene therapy, have recently been investigated in tumor model systems to provide a more complete understanding of hepatocarcinogenesis and effectively design therapeutic strategies to treat this disease. In our study, we constructed an adenoviral vector expressing small interfering RNA (siRNA) targeting a newly discovered gene named upregulated gene 11 (URG11). We introduced this vector into HCC cells to investigate the role of URG11 in HCC carcinogenesis. We observed that upon URG11 knockdown, HCC cell proliferation was inhibited through downregulation of several G1-S phase related molecules including cyclin D1 and apoptosis was induced as a result of Bcl-2 downregulation. Besides decreased expression of cyclin D1, CDK4, pRb and Bcl-2, URG11 also suppressed several other proteins including CAPN9, which was identified by cDNA microarray and 2D gel electrophoresis. Moreover, Ad-URG11-siRNA significantly suppressed HCC tumor growth in nude mice. In conclusion, Ad-URG11-siRNA can significantly suppress HCC tumor growth in vitro and in vivo by silencing the URG11 gene, and the use of this vector for gene therapy may represent a novel strategy to treat human HCC.


Subject(s)
Adenoviridae/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , RNA Interference , Trans-Activators/genetics , Carcinoma, Hepatocellular/genetics , Cell Division/genetics , Cell Line, Tumor , Cell Proliferation , Humans , Liver Neoplasms/genetics
11.
Oncotarget ; 12(24): 2421-2433, 2021 Nov 23.
Article in English | MEDLINE | ID: mdl-34853663

ABSTRACT

Hepatitis B virus (HBV) is a human pathogen that has infected an estimated two billion people worldwide. Despite the availability of highly efficacious vaccines, universal screening of the blood supply for virus, and potent direct acting anti-viral drugs, there are more than 250 million carriers of HBV who are at risk for the sequential development of hepatitis, fibrosis, cirrhosis and hepatocellular carcinoma (HCC). More than 800,000 deaths per year are attributed to chronic hepatitis B. Many different therapeutic approaches have been developed to block virus replication, and although effective, none are curative. These treatments have little or no impact upon the portions of integrated HBV DNA, which often encode the virus regulatory protein, HBx. Although given little attention, HBx is an important therapeutic target because it contributes importantly to (a) HBV replication, (b) in protecting infected cells from immune mediated destruction during chronic infection, and (c) in the development of HCC. Thus, the development of therapies targeting HBx, combined with other established therapies, will provide a functional cure that will target virus replication and further reduce or eliminate both the morbidity and mortality associated with chronic liver disease and HCC. Simultaneous targeting of all these characteristics underscores the importance of developing therapies against HBx.

12.
Viruses ; 13(1)2021 Jan 15.
Article in English | MEDLINE | ID: mdl-33467678

ABSTRACT

While treatment options are available for hepatitis B virus (HBV), there is currently no cure. Anti-HBV nucleoside analogs and interferon-alpha 2b rarely clear HBV covalently closed circular DNA (cccDNA), requiring lifelong treatment. Recently, we identified GLP-26, a glyoxamide derivative which modulates HBV capsid assembly. The impact of GLP-26 on viral replication and integrated DNA was assessed in an HBV nude mouse model bearing HBV transfected AD38 xenografts. At day 45 post-infection, GLP-26 reduced HBV titers by 2.3-3 log10 versus infected placebo-treated mice. Combination therapy with GLP-26 and entecavir reduced HBV log10 titers by 4.6-fold versus placebo. Next, we examined the pharmacokinetics (PK) in cynomolgus monkeys administered GLP-26 via IV (1 mg/kg) or PO (5 mg/kg). GLP-26 was found to have 34% oral bioavailability, with a mean input time of 3.17 h. The oral dose produced a mean peak plasma concentration of 380.7 ng/mL, observed 0.67 h after administration (~30-fold > in vitro EC90 corrected for protein binding), with a mean terminal elimination half-life of 2.4 h and a mean area under the plasma concentration versus time curve of 1660 ngĀ·hr/mL. GLP-26 was 86.7% bound in monkey plasma. Lastly, GLP-26 demonstrated a favorable toxicity profile confirmed in primary human cardiomyocytes. Thus, GLP-26 warrants further preclinical development as an add on to treatment for HBV infection.


Subject(s)
Capsid/drug effects , Capsid/metabolism , Cardiotoxins/pharmacokinetics , Hepatitis B virus/drug effects , Hepatitis B virus/physiology , Sulfonylurea Compounds/pharmacokinetics , Virus Assembly/drug effects , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Hepatitis B/drug therapy , Hepatitis B/virology , Humans , Macaca fascicularis , Male , Mice , Myocytes, Cardiac/drug effects , Sulfonylurea Compounds/adverse effects , Sulfonylurea Compounds/chemistry , Viral Load
13.
Int J Cancer ; 124(12): 2886-92, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19253371

ABSTRACT

Hepatitis C virus (HCV) infection is often associated with chronic liver disease, which is a major risk factor for the development of hepatocellular carcinoma (HCC). To study the HCV host-cell relationship on the molecular level, HepG2 and Huh7 cells were stably transfected with an infectious cDNA clone of HCV or with empty vector. Evidence for HCV replication was obtained in both culture systems. HCV also stimulated growth in vitro. To identify genes whose altered expression by HCV are important to the pathogenesis of infection, RNAs were isolated from HepG2-HCV and HepG2-vector cells and subjected to microarray analysis. The results showed that arginase 1 mRNA and protein were elevated about threefold in HCV positive compared with negative cells (p < 0.01). Arginase 1 expression was elevated in more than 75% of HCV infected liver samples compared with paired HCC from the same patients (>33% positive) and to uninfected liver tissues (0% positive). Arginase 1 specific siRNA inhibited the ability of HCV to stimulate hepatocellular growth in culture by >70%, suggesting that the metabolism of arginine to ornithine may contribute to HCV mediated stimulation of hepatocellular growth. Introduction of arginase specific siRNA also resulted in increased nitric oxide synthase (iNOS) (>1.2-fold), nitric oxide (NO) production (>3-fold) and increased cell death (>2.5-fold) in HCV positive compared with negative cells, suggesting that these molecules potentially contribute to hepatocellular damage. Hence, an important part of the mechanism whereby HCV regulates hepatocellular growth and survival may be through altering arginine metabolism.


Subject(s)
Arginase/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/physiology , Hepacivirus/physiology , Hepatitis C/complications , Liver Neoplasms/genetics , Blotting, Western , Carcinoma, Hepatocellular/virology , DNA, Viral/administration & dosage , Genetic Vectors , Hepatitis C/enzymology , Hepatitis C/pathology , Humans , Liver Neoplasms/virology , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transfection , Tumor Cells, Cultured , Virus Replication
14.
Int J Cancer ; 125(8): 1894-901, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19610068

ABSTRACT

Hepatitis B x antigen, or HBxAg, contributes importantly to the pathogenesis of hepatocellular carcinoma (HCC). Given that HBxAg constitutively activates beta-catenin and that upregulated ErbB-2 promotes beta-catenin signaling in other tumor types, experiments were designed to ask whether HBxAg was associated with upregulated expression of ErbB-2. When HBxAg positive and negative HepG2 cells were subjected to proteomics analysis, ErbB-2 was shown to be upregulated in HepG2X but not control cells. ErbB-2 was also strongly upregulated in HB infected liver, and weakly in some HCC nodules, where it correlated with HBxAg expression. Among tumor bearing patients, strong ErbB-2 staining in the liver was associated with dysplasia, and a shorter survival after tumor diagnosis. This implies that elevated ErbB-2 is an early marker of HCC. Treatment of HepG2X cells with ErbB-2 specific siRNA not only reduced ErbB-2 expression, but also reduced the expression of beta-catenin, suggesting that ErbB-2 contributed to the stabilization of beta-catenin. ErbB-2 specific siRNA also partially blocked the ability of HBxAg to promote DNA synthesis and growth of HepG2 cells. These results suggest that ErbB-2/beta-catenin up-regulation contributes importantly to the mechanism of HBxAg mediated hepatocellular growth.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/mortality , Liver Neoplasms/metabolism , Liver Neoplasms/mortality , Liver/metabolism , Receptor, ErbB-2/metabolism , Trans-Activators/metabolism , Adult , Blotting, Western , Carcinoma, Hepatocellular/secondary , Cell Proliferation , Female , Follow-Up Studies , Hepatitis, Chronic/metabolism , Hepatitis, Chronic/pathology , Humans , Immunoenzyme Techniques , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Retrospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Viral Regulatory and Accessory Proteins , beta Catenin/metabolism
15.
Mol Carcinog ; 48(1): 56-65, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18506760

ABSTRACT

Hepatitis B virus (HBV) X protein (HBx) and cyclooxygenase-2 (COX-2) are all playing roles in hepatocellular carcinoma (HCC), but the reversing effects of COX-2 inhibitors on the neoplastic features caused by HBx protein is still unclear. To further evaluate the therapeutic potential of celecoxib on HBx mediated transformation, HCC cells transfected with HBx gene were treated with COX-2 selective inhibitor, celecoxib. The amount the main metabolite of COX-2, prostaglandin E2 (PGE2), was determined by using high sensitivity ELISA. Electron microscope and flow cytometry was used to analyze cell apoptosis and cell cycle distribution. RT-PCR and Western blot were used to identify the molecules involved in celecoxib induced cell apoptosis. The results showed that celecoxib inhibited cell growth more significantly and also induced more cell apoptosis in HBx over-expression cells than in control cells. Celecoxib could selectively inhibited COX-2 expression and PGE2 production. Celecoxib also inhibited p(473Ser)Akt, raf and p53 expression, and induced apoptosis by release of cytochrome c and activation of caspase 9, 3, and 6, which were more remarkably in HBx positive cells than in control cells. These results suggest that celecoxib had potent cell growth inhibitory effects on HBx positive HCC cells mainly through inducing more cell apoptosis, and these findings provide a new insight into the anticancer effects of celecoxib against HBx related HCC.


Subject(s)
Apoptosis/drug effects , Carcinoma, Hepatocellular/pathology , Cyclooxygenase 2 Inhibitors/pharmacology , Liver Neoplasms/pathology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Trans-Activators/physiology , Viral Regulatory and Accessory Proteins/physiology , Blotting, Western , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/virology , Caspase Inhibitors , Caspases/metabolism , Celecoxib , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cyclooxygenase 2/chemistry , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoblotting , Immunoenzyme Techniques , Immunoprecipitation , Liver Neoplasms/enzymology , Liver Neoplasms/virology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-raf/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
16.
Carcinogenesis ; 29(6): 1207-14, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18477650

ABSTRACT

BACKGROUND: The hepatitis B virus x gene (HBx) is a promiscuous transactivator implicated in the development of hepatocellular carcinoma (HCC). The present study was designed to investigate the molecular events regulated by HBx. METHODS: Genomic and proteomic expression profiling was performed in Huh7 HCC cells transfected with HBx mutants with a C-terminal deletion. The gene and protein expression of wingless-type murine-mammary-tumour virus (MMTV) integration site family, member 5A (Wnt-5a) was validated by analyses of reverse transcription-polymerase chain reaction (RT-PCR), real-time RT-PCR, western blot and immunohistochemistry. RESULTS: Differentially expressed genes and proteins were found in the transfected Huh7 HCC cells; most of them were involved in transcriptional regulation, although others including oncogenes or tumor suppressor genes, and molecules involved in cell junctions, signal transduction pathways, metabolism or the immune response were also observed. The expression of the Wnt-5a gene was elevated >10-fold in Huh7 cells transfected with the HBx3'-30 amino acid deletion mutant. However, the expression was downregulated by the transfection with the HBx3'-40 amino acid deletion mutant. The changes in Wnt-5a expression were also observed in human HCC tissues, compared with corresponding non-cancerous liver tissues. A negative correlation was found between the expression of Wnt-5a and HBx COOH mutations in HCC tissues. CONCLUSIONS: HBx mutants may participate in the development and progression of HCC, at least in part through the Wnt-5a pathway.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Liver Neoplasms/genetics , Liver Neoplasms/virology , Proto-Oncogene Proteins/biosynthesis , Trans-Activators/genetics , Wnt Proteins/biosynthesis , Blotting, Western , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Gene Expression , Humans , Immunohistochemistry , Liver Neoplasms/metabolism , Mutation , Oligonucleotide Array Sequence Analysis , Proteomics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Viral Regulatory and Accessory Proteins , Wnt-5a Protein
17.
Biochem Biophys Res Commun ; 373(4): 643-7, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18602370

ABSTRACT

The role of the hepatitis B virus X protein (HBx) in hepatocarcinogenesis remains controversial. To investigate the biological impact of hepatitis B virus x gene (HBx) mutation on hepatoma cells, plasmids expressing the full-length HBx or HBx deletion mutants were constructed. The biological activities in these transfectants were analyzed by a series of assays. Results showed that HBx3'-20 and HBx3'-40 amino acid deletion mutants exhibited an increase in cellular proliferation, focus formation, tumorigenicity, and invasive growth and metastasis through promotion of the cell cycle from G0/G1 to the S phase, when compared with the full-length HBx. In contrast, HBx3'-30 amino acid deletion mutant repressed cell proliferation by blocking in G1 phase. The expression of P53, p21(WAF1), p14(ARF), and MDM2 proteins was regulated by expression of HBx mutants. In conclusions, HBx variants showed different effects and functions on cell proliferation and invasion by regulation of the cell cycle progression and its associated proteins expression.


Subject(s)
Carcinoma, Hepatocellular/virology , Cell Transformation, Viral , Hepatitis B virus/metabolism , Liver Neoplasms/virology , Trans-Activators/metabolism , Apoptosis , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Hepatitis B virus/genetics , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Neoplasm Invasiveness , Plasmids , Proto-Oncogene Proteins c-mdm2/metabolism , Sequence Deletion , Trans-Activators/genetics , Transfection , Tumor Suppressor Protein p14ARF/metabolism , Tumor Suppressor Protein p53/metabolism , Viral Regulatory and Accessory Proteins
18.
World J Gastroenterol ; 14(9): 1346-52, 2008 Mar 07.
Article in English | MEDLINE | ID: mdl-18322946

ABSTRACT

AIM: To investigate the hepatitis B virus (HBV)x gene (HBx) state in the tissues of HBV-related hepatocellular carcinoma (HCC) in Chinese patients and whether there were particular HBx mutations. METHODS: HBx gene was amplified and direct sequencing was used in genomic DNA samples from 20 HCC and corresponding non-cancerous liver tissues from HBsAg-positive patients. HBV DNA integration and HBx deleted mutation were validated in 45 HCC patients at different stages by Southern blot analysis and polymerase chain reaction methods. RESULTS: The frequencies of HBx point mutations were significantly lower in HCC than their corresponding non-cancerous liver tissues (11/19 vs 18/19, P=0.019). In contrast, deletions in HBx gene were significantly higher in HCC than their non-cancerous liver tissues (16/19 vs 4/19, P<0.001). The deletion of HBx COOH-terminal was detected in 14 HCC tissues. A specific integration of HBx at 17p13 locus was also found in 8 of 16 HCC, and all of them also exhibited full-length HBx deletions. Integrated or integrated coexistence with replicated pattern was obtained in 45.5% (20/45)-56.8% (25/45) tumors and 40.9% (18/45)-52.3% (23/45) non-tumor tissues. CONCLUSION: HBx deletion, especially the COOH-terminal deletion of HBx is a frequent event in HBV-associated HCC tissues in China. HBV integration had also taken place in partial HCC tissues. This supporting the hypothesis that deletion and probably integrated forms of the HBx gene may be implicated in liver carcinogenesis.


Subject(s)
Carcinoma, Hepatocellular/virology , Gene Deletion , Liver Neoplasms/virology , Viral Regulatory and Accessory Proteins/genetics , Adult , Aged , Amino Acid Sequence , Base Sequence , Carcinoma, Hepatocellular/ethnology , Carcinoma, Hepatocellular/genetics , Case-Control Studies , China , DNA, Neoplasm/genetics , DNA, Viral/genetics , Female , Gene Amplification , Hepatitis B Surface Antigens/metabolism , Hepatitis B virus/genetics , Hepatitis B virus/immunology , Humans , Liver Neoplasms/ethnology , Liver Neoplasms/genetics , Male , Middle Aged , Molecular Sequence Data , Point Mutation/genetics , Trans-Activators
20.
Cancer Lett ; 252(2): 157-70, 2007 Jul 18.
Article in English | MEDLINE | ID: mdl-17188425

ABSTRACT

Chronic liver disease associated with long term hepatitis B virus (HBV) infection contributes importantly to the development of hepatocellular carcinoma (HCC). A salient feature of these chronic infections is the integration of subgenomic HBV DNA fragments into many different locations within the host DNA, suggesting that integration is random. Although this may promote genetic instability during liver regeneration which accompanies a bout of chronic liver disease, the actual role of integrated HBV DNA in hepatocarcinogenesis is uncertain. Importantly, most integration events retain the HBV open reading frame encoding the HBx antigen (HBxAg), which is the virus contribution to HCC. In addition, many integration events reported in the literature occur near or within fragile sites or other cancer associated regions of the human genome that are prone to instability in tumor development and progression. Genetic instability associated with integration potentially alters the expression of oncogenes, tumor suppressor genes, and microRNAs (miRNAs) that may contribute importantly to tumorigenesis. If so, then selected integration events may alter pathways that are rate limiting in hepatocarcinogenesis, thereby providing targets with diagnostic/prognostic potential and for therapeutic intervention.


Subject(s)
Cell Transformation, Neoplastic , Chromosome Fragile Sites , Hepatitis B virus/physiology , Liver Neoplasms/virology , Virus Integration , Humans , Liver Neoplasms/genetics , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL