Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Respirol Case Rep ; 12(6): e01418, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38903946

ABSTRACT

Amyloidosis is a pathological deposition disease that causes a spectrum of organ dysfunction. Pulmonary involvement is generally associated with immunoglobulin light chain type (AL) amyloid. Transthyretin (ATTR) amyloid build up in the lung is thought to be a senile disease observed usually as a finding at autopsy. We describe a case of pulmonary ATTR amyloidosis with concurrent mycobacterial tuberculosis infection.

2.
Med Mycol Case Rep ; 34: 22-26, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34584835

ABSTRACT

We report the first published case of Prototheca wickerhamii breast implant infection. This occurred after mastectomy, chemotherapy, radiotherapy, breast reconstruction, implant revisions and breast seroma aspirations and was preceded by polymicrobial infection. Definitive treatment required implant removal and intravenous liposomal amphotericin B. The management of breast prosthesis infections is discussed.

4.
PLoS One ; 10(9): e0138522, 2015.
Article in English | MEDLINE | ID: mdl-26390404

ABSTRACT

INTRODUCTION: Immunosuppressants are used ubiquitously post-liver transplantation to prevent allograft rejection. However their effects on hepatocytes are unknown. Experimental data from non-liver cells indicate that immunosuppressants may promote cell death thereby driving an inflammatory response that promotes fibrosis and raises concerns that a similar effect may occur within the liver. We evaluated apoptosis within the liver tissue of post-liver transplant patients and correlated these findings with in vitro experiments investigating the effects of immunosuppressants on apoptosis in primary hepatocytes. METHODS: Hepatocyte apoptosis was assessed using immunohistochemistry for M30 CytoDEATH and cleaved PARP in human liver tissue. Primary mouse hepatocytes were treated with various combinations of cyclosporine, tacrolimus, sirolimus, or MMF. Cell viability and apoptosis were evaluated using crystal violet assays and Western immunoblots probed for cleaved PARP and cleaved caspase 3. RESULTS: Post-liver transplant patients had a 4.9-fold and 1.7-fold increase in M30 CytoDEATH and cleaved PARP compared to normal subjects. Cyclosporine and tacrolimus at therapeutic concentrations did not affect hepatocyte apoptosis, however when they were combined with MMF, cell death was significantly enhanced. Cell viability was reduced by 46% and 41%, cleaved PARP was increased 2.6-fold and 2.2-fold, and cleaved caspase 3 increased 2.2-fold and 1.8-fold following treatment with Cyclosporine/MMF and Tacrolimus/MMF respectively. By contrast, the sirolimus/MMF combination did not significantly reduce hepatocyte viability or promote apoptosis. CONCLUSION: Commonly used immunosuppressive drug regimens employed after liver transplantation enhance hepatocyte cell death and may thus contribute to the increased liver fibrosis that occurs in a proportion of liver transplant recipients.


Subject(s)
Apoptosis/drug effects , Hepatocytes/drug effects , Immunosuppressive Agents/therapeutic use , Liver Transplantation/adverse effects , Liver/drug effects , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Caspase 3/metabolism , Cell Survival/drug effects , Cyclosporine/pharmacology , Cyclosporine/urine , Drug Therapy, Combination/methods , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Hepatocytes/metabolism , Humans , Immunosuppressive Agents/pharmacology , Liver/metabolism , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Transplantation/methods , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mitomycin/pharmacology , Mitomycin/therapeutic use , Semustine/pharmacology , Semustine/therapeutic use , Sirolimus/pharmacology , Sirolimus/therapeutic use , Tacrolimus/pharmacology , Tacrolimus/therapeutic use
5.
Mol Ther ; 23(9): 1434-43, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25997428

ABSTRACT

Angiotensin converting enzyme 2 (ACE2) which breaks down profibrotic peptide angiotensin II to antifibrotic peptide angiotensin-(1-7) is a potential therapeutic target in liver fibrosis. We therefore investigated the long-term therapeutic effect of recombinant ACE2 using a liver-specific adeno-associated viral genome 2 serotype 8 vector (rAAV2/8-ACE2) with a liver-specific promoter in three murine models of chronic liver disease, including carbon tetrachloride-induced toxic injury, bile duct ligation-induced cholestatic injury, and methionine- and choline-deficient diet-induced steatotic injury. A single injection of rAAV2/8-ACE2 was administered after liver disease has established. Hepatic fibrosis, gene and protein expression, and the mechanisms that rAAV2/8-ACE2 therapy associated reduction in liver fibrosis were analyzed. Compared with control group, rAAV2/8-ACE2 therapy produced rapid and sustained upregulation of hepatic ACE2, resulting in a profound reduction in fibrosis and profibrotic markers in all diseased models. These changes were accompanied by reduction in hepatic angiotensin II levels with concomitant increases in hepatic angiotensin-(1-7) levels, resulting in significant reductions of NADPH oxidase assembly, oxidative stress and ERK1/2 and p38 phosphorylation. Moreover, rAAV2/8-ACE2 therapy normalized increased intrahepatic vascular tone in fibrotic livers. We conclude that rAAV2/8-ACE2 is an effective liver-targeted, long-term therapy for liver fibrosis and its complications without producing unwanted systemic effects.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Genetic Vectors/genetics , Liver Cirrhosis/genetics , Liver Cirrhosis/metabolism , Peptidyl-Dipeptidase A/genetics , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Cytokines/metabolism , Dependovirus/classification , Disease Models, Animal , Enzyme Activation , Gene Expression , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Hepatic Stellate Cells/metabolism , Inflammation Mediators/metabolism , Injections, Intraperitoneal , Lipid Peroxidation/genetics , Liver Cirrhosis/chemically induced , Liver Cirrhosis/pathology , Liver Cirrhosis/therapy , Liver Function Tests , MAP Kinase Signaling System , Male , Methoxamine/pharmacology , Mice , NADPH Oxidases/metabolism , Neovascularization, Pathologic/genetics , Organ Specificity/genetics , Oxidative Stress , Peptidyl-Dipeptidase A/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Proc Natl Acad Sci U S A ; 112(18): 5797-802, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25902529

ABSTRACT

Hepatitis B virus (HBV) infection can result in a spectrum of outcomes from immune-mediated control to disease progression, cirrhosis, and liver cancer. The host molecular pathways that influence and contribute to these outcomes need to be defined. Using an immunocompetent mouse model of chronic HBV infection, we identified some of the host cellular and molecular factors that impact on infection outcomes. Here, we show that cellular inhibitor of apoptosis proteins (cIAPs) attenuate TNF signaling during hepatitis B infection, and they restrict the death of infected hepatocytes, thus allowing viral persistence. Animals with a liver-specific cIAP1 and total cIAP2 deficiency efficiently control HBV infection compared with WT mice. This phenotype was partly recapitulated in mice that were deficient in cIAP2 alone. These results indicate that antagonizing the function of cIAPs may promote the clearance of HBV infection.


Subject(s)
Hepatitis B virus , Hepatitis B/metabolism , Inhibitor of Apoptosis Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Baculoviral IAP Repeat-Containing 3 Protein , CD4 Antigens/metabolism , CD8 Antigens/metabolism , Cytokines/metabolism , DNA, Viral/genetics , Disease Models, Animal , Genotype , Hepatocytes/metabolism , Hepatocytes/virology , Immunophenotyping , Immunosuppression Therapy , Interferon-gamma/metabolism , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/virology , Mice , Mice, Inbred C57BL , Phenotype , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
7.
J Gen Virol ; 95(Pt 10): 2204-2215, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24973240

ABSTRACT

Chronic hepatitis C virus (HCV) infection results in progressive liver fibrosis leading to cirrhosis and liver cancer. The mechanism for this remains unclear but hepatocyte apoptosis is thought to play a major role. Hepatocyte apoptosis in human liver tissue was determined by immunohistochemistry for cytokeratin 18 (M30 CytoDEATH) and cleaved poly(ADP-ribose) polymerase (PARP). In vitro studies were performed with replication-defective recombinant adenoviruses expressing HCV proteins (rAdHCV) to study the effects of HCV on cell death in Huh7 cells, primary mouse hepatocytes (PMoHs) and primary human hepatocytes (PHHs). Cell viability and apoptosis were studied using crystal violet assays and Western blots probed for cleaved caspase-3 and cleaved PARP, with and without treatment with the pan-caspase inhibitor Q-VD-OPh and necrostatin-1. Liver tissue of HCV-infected patients expressed elevated levels of apoptotic markers compared with HCV-negative patients. rAdHCV infection reduced cell viability compared with uninfected controls and cells infected with control virus (rAdGFP). Huh7, PMoHs and PHHs infected with rAdHCV showed significantly increased levels of apoptotic markers compared with uninfected controls and rAdGFP-infected cells. In rAdHCV-infected Huh7, treatment with Q-VD-OPh and necrostatin-1 both improved cell viability. Q-VD-Oph also reduced cleaved PARP in rAdHCV-infected Huh7 and PMoHs. Hepatocyte apoptosis is known to be increased in the livers of HCV-infected patients. HCV promoted cell death in primary and immortalized hepatocytes, and this was inhibited by Q-VD-OPh and necrostatin-1. These findings indicate that HCV-induced cell death occurs by both apoptosis and necroptosis, and provide new insights into the mechanisms of HCV-induced liver injury.


Subject(s)
Apoptosis , Hepacivirus/physiology , Hepatitis C/pathology , Hepatocytes/physiology , Hepatocytes/virology , Necrosis , Amino Acid Chloromethyl Ketones/metabolism , Animals , Caspases/analysis , Cell Survival , Enzyme Inhibitors/metabolism , Hepatocytes/drug effects , Humans , Imidazoles/metabolism , Indoles/metabolism , Mice , Mice, Inbred C57BL , Quinolines/metabolism
8.
J Clin Virol ; 56(3): 226-31, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23218952

ABSTRACT

BACKGROUND: Infection with pandemic A/H1N1/2009 influenza virus led to hospitalisation of patients not expected to be at risk of severe disease from seasonal influenza infection. OBJECTIVES: We sought to establish whether (i) DC maturation was compromised in patients experiencing severe pandemic influenza infection, (ii) the pandemic virus differed from seasonal influenza virus in its ability to induce DC maturation and (iii) there was an associated inability to activate memory B cells or induce antibody. STUDY DESIGN: Peripheral blood mononuclear (PBMCs) cells were sampled from individuals with confirmed acute pandemic A/H1N1/2009 influenza infection or from healthy vaccinated controls. DCs were differentiated from the PBMC and tested for their ability to mature following stimulation with pandemic virus, seasonal H3N2 influenza virus or LPS. Serum samples from the patients were used to assess seroconversion to influenza and the levels of influenza specific memory B cells in PBMC were also determined. RESULTS: DCs obtained from all individuals exhibited negligible maturation marker upregulation when exposed to pandemic A/H1N1/2009 virus but showed a strong response to the seasonal H3N2 virus and LPS. Robust levels of memory B cell were obtained in both groups and patients seroconverted to the virus. CONCLUSIONS: Overall, the ability of patient's DC to mature in response to different stimuli was no different to that of control subjects DCs. Importantly, panH1N109 virus failed to induce substantial DC maturation in any individual, contrasting with seasonal virus, but this did not result in failure to mount memory B cell and antibody responses to the virus.


Subject(s)
Dendritic Cells/immunology , Influenza A Virus, H1N1 Subtype/immunology , Influenza, Human/immunology , Influenza, Human/virology , Antibodies, Viral/immunology , B-Lymphocytes/immunology , Cell Differentiation , Humans , Immune Evasion , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza A Virus, H3N2 Subtype/immunology , Leukocytes, Mononuclear/immunology , Lipopolysaccharides/immunology
9.
Infect Dis Ther ; 2(2): 145-58, 2013 Dec.
Article in English | MEDLINE | ID: mdl-25134477

ABSTRACT

The Japanese encephalitis virus (JEV) is endemic in many countries in southern Asia and the western Pacific Rim, with new spread to previously unrecognized countries. It is an important cause of childhood neurological disease associated with permanent neurological sequelae and death. Fortunately, JE is a vaccine-preventable disease. The ChimeriVax™-JE (Sanofi Pasteur, Lyon, France) is a live-attenuated chimeric vaccine derived from the live-attenuated yellow fever virus, YF17D, which expresses the envelope proteins of the attenuated JEV vaccine strain, SA14-14-2. It is a safe, well-tolerated vaccine that is highly immunogenic in adults and children. The average geometric mean neutralizing antibody titer (GMT) in adults is 1,392 and over 90% of adults remain seroprotected 5 years after vaccination. In children and toddlers, more than 80% remain seroprotected 2 years after primary vaccination and demonstrate a robust and durable anamnestic response (>500-fold rise in GMT) with 99.1% seroprotection rates 1 year after a booster vaccine dose. The ChimeriVax™-JE is effective in children living in endemic regions where the vaccine could possibly be integrated into existing childhood vaccination programs. ChimeriVax™-JE is also indicated for travelers at risk of JE infection.

10.
PLoS One ; 7(10): e47492, 2012.
Article in English | MEDLINE | ID: mdl-23091628

ABSTRACT

Although many studies provide strong evidence supporting the development of HCV virus-like particle (VLP)-based vaccines, the fact that heterologous viral vectors and/or multiple dosing regimes are required to induce protective immunity indicates that it is necessary to improve their immunogenicity. In this study, we have evaluated the use of an anionic self-adjuvanting lipopeptide containing the TLR2 agonist Pam(2)Cys (E(8)Pam(2)Cys) to enhance the immunogenicity of VLPs containing the HCV structural proteins (core, E1 and E2) of genotype 1a. While co-formulation of this lipopeptide with VLPs only resulted in marginal improvements in dendritic cell (DC) uptake, its ability to concomitantly induce DC maturation at very small doses is a feature not observed using VLPs alone or in the presence of an aluminium hydroxide-based adjuvant (Alum). Dramatically improved VLP and E2-specific antibody responses were observed in VLP+E(8)Pam(2)Cys vaccinated mice where up to 3 doses of non-adjuvanted or traditionally alum-adjuvanted VLPs was required to match the antibody titres obtained with a single dose of VLPs formulated with this lipopeptide. This result also correlated with significantly higher numbers of specific antibody secreting cells that was detected in the spleens of VLP+E(8)Pam(2)Cys vaccinated mice and greater ability of sera from these mice to neutralise the binding and uptake of VLPs by Huh7 cells. Moreover, vaccination of HLA-A2 transgenic mice with this formulation also induced better VLP-specific IFN-γ-mediated responses compared to non-adjuvanted VLPs but comparable levels to that achieved when coadministered with complete freund's adjuvant. These results suggest overall that the immunogenicity of HCV VLPs can be significantly improved by the addition of this novel adjuvant by targeting their delivery to DCs and could therefore constitute a viable vaccine strategy for the treatment of HCV.


Subject(s)
Antibody Formation/immunology , Dendritic Cells/immunology , Hepatitis C/immunology , Immunity, Cellular/immunology , Lipopeptides/immunology , Toll-Like Receptor 2/chemistry , Vaccines, Virus-Like Particle/immunology , Animals , Antibodies, Neutralizing/immunology , Cell Line , Dendritic Cells/metabolism , Female , Hepatitis C/genetics , Hepatitis C Antibodies/immunology , Humans , Mice , Vaccines, Virus-Like Particle/genetics , Virus Internalization
11.
World J Gastroenterol ; 18(18): 2172-9, 2012 May 14.
Article in English | MEDLINE | ID: mdl-22611309

ABSTRACT

Hepatitis C (HCV)-infected patients have a poorer survival post-liver transplantation compared to patients transplanted for other indications, since HCV recurrence post-transplant is universal and commonly follows an aggressive course. There is increasing evidence that in the non-transplant setting, induction of hepatocyte apoptosis is one of the main mechanisms by which HCV drives liver inflammation and fibrosis, and that HCV proteins directly promote apoptosis. Recent studies have shown that post-liver transplant, there is a link between high levels of HCV replication, enhanced hepatocyte apoptosis and the subsequent development of rapidly progressive liver fibrosis. Although the responsible mechanisms remain unclear, it is likely that immunosuppressive drugs play an important role. It is well known that immunosuppressants impair immune control of HCV, thereby allowing increased viral replication. However there is also evidence that immunosuppressants may directly induce apoptosis and this may be facilitated by the presence of high levels of HCV replication. Thus HCV and immunosuppressants may synergistically interact to further enhance apoptosis and drive more rapid fibrosis. These findings suggest that modulation of apoptosis within the liver either by changing immunosuppressive therapy or the use of apoptosis inhibitors may help prevent fibrosis progression in patients with post-transplant HCV disease.


Subject(s)
Apoptosis/drug effects , Hepacivirus/pathogenicity , Hepatitis C/surgery , Immunosuppressive Agents/adverse effects , Liver Transplantation/adverse effects , Liver/drug effects , Liver/virology , Animals , Hepatitis C/complications , Hepatitis C/diagnosis , Humans , Liver/pathology , Liver Cirrhosis/pathology , Liver Cirrhosis/virology , Recurrence , Risk Factors , Treatment Outcome , Virus Replication
12.
J Hepatol ; 53(4): 599-607, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20667615

ABSTRACT

BACKGROUND & AIMS: HCV patients who fail conventional interferon-based therapy have limited treatment options. Dendritic cells are central to the priming and development of antigen-specific CD4(+) and CD8(+) T cell immunity, necessary to elicit effective viral clearance. The aim of the study was to investigate the safety and efficacy of vaccination with autologous dendritic cells loaded with HCV-specific cytotoxic T cell epitopes. METHODS: We examined the potential of autologous monocyte-derived dendritic cells (MoDC), presenting HCV-specific HLA A2.1-restricted cytotoxic T cell epitopes, to influence the course of infection in six patients who failed conventional therapy. Dendritic cells were loaded and activated ex vivo with lipopeptides. In this phase 1 dose escalation study, all patients received a standard dose of cells by the intradermal route while sequential patients received an increased dose by the intravenous route. RESULTS: No patient showed a severe adverse reaction although all experienced transient minor side effects. HCV-specific CD8(+) T cell responses were enumerated in PBMC by ELIspot for interferon-gamma. Patients generated de novo responses, not only to peptides presented by the cellular vaccine but also to additional viral epitopes not represented in the lipopeptides, suggestive of epitope spreading. Despite this, no increases in ALT levels were observed. However, the responses were not sustained and failed to influence the viral load, the anti-HCV core antibody response and the level of circulating cytokines. CONCLUSIONS: Immunotherapy using autologous MoDC pulsed with lipopeptides was safe, but was unable to generate sustained responses or alter the outcome of the infection. Alternative dosing regimens or vaccination routes may need to be considered to achieve therapeutic benefit.


Subject(s)
Dendritic Cells/immunology , Hepatitis C, Chronic/immunology , Hepatitis C, Chronic/prevention & control , Vaccination , Adolescent , Adult , Aged , Female , Humans , Male , Middle Aged , Treatment Outcome
13.
Virus Res ; 147(1): 7-16, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19786052

ABSTRACT

BACKGROUND/AIMS: Dysregulation of the cell cycle is frequently associated with tumor development. Hepatitis B virus (HBV) is associated with a significant risk of developing hepatocellular carcinoma but the effects of HBV on cell cycle regulation are not completely understood. METHODS: We have used a recombinant adeno-HBV model system to investigate the effect of infection with HBV and the replication defective lamivudine resistant mutant rtM204I mutant on hepatocyte cell cycle and cell viability. RESULTS: Huh7 cells synchronised at the G1/S phase of the cell cycle were arrested at the G2/M following infection with rAdHBV-wt and rAdHBV-M204I. This was accompanied by increased levels of p21(cip1), p-cdc2, cyclins D, A and B. Cell viability was reduced and cleaved caspase 3 levels were increased in HBV- and rtM204I-infected cells. rAdHBV-M204I-infected Huh7 cells also demonstrated significant up-regulation of phospho-ERK, phospho-Akt, p53 and phospho-Mdm2 compared to mock-infected cells. These changes were comparable to those following infection of Huh7 cells with rAdHBV-wt. CONCLUSION: Our results suggest that HBV, regardless of phenotype, produces cell cycle arrest and reduced hepatocyte viability. Perturbations in these cellular processes are likely to underlie HBV-associated liver oncogenic transformation and may help explain the ongoing risk of developing hepatocellular carcinoma in individuals in whom the lamivudine resistant rtM204I mutant emerges.


Subject(s)
Cell Cycle , Hepatitis B virus/pathogenicity , Hepatocytes/physiology , Hepatocytes/virology , Cell Cycle Proteins/analysis , Cell Line , Cell Survival , Hepatocytes/chemistry , Humans , Protein Kinases/analysis , Signal Transduction
14.
Virus Res ; 148(1-2): 51-9, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005910

ABSTRACT

The mechanism by which hepatitis B virus (HBV) infection causes severe inflammatory liver diseases is multifactorial and related to interactions with cell signaling pathways and the ensuing inflammatory response. Activation of JAK/STAT/SOCS signaling is essential for the induction of cellular antiviral responses, contributes to apoptosis and is negatively regulated by SOCS proteins. Recent reports have shown that SOCS3 activation interferes with viral protein expression and treatment response and thereby plays a major role in hepatitis virus infections. We analyzed the expression of SOCS3 in liver specimens from HBV-infected patients using immunohistochemistry (IHC) and determined the effect of HBV on STAT/SOCS signaling in functional cell culture experiments (HuH-7) using HBV-expressing adenoviral constructs (AdHBV). Increased expression of SOCS3 protein was identified in liver specimens from patients with chronic HBV-infection and this correlated with the severity of liver inflammation. In accordance with the IHC-findings, in vitro analyses demonstrated that HBV infection of HuH7 cells was associated with increased expression of SOCS3 protein. In spite of the over expression of its negative regulator SOCS3 we observed a constitutive activation of STAT3. SOCS1 levels were not increased while pSTAT1 was suppressed in HBV-infected HuH7 cells. Our results demonstrate that STAT/SOCS-signaling is dysregulated in HBV-infected hepatocytes both in vivo and in vitro and this correlated with the severity of liver inflammatory changes. This interference of STAT/SOCS signaling by HBV may result in an ineffective immune response against HBV and potentially contributes to viral pathogenesis, malignant transformation and may represent an important mechanism of viral persistence.


Subject(s)
Hepatitis B virus/physiology , Hepatitis B/immunology , Liver/immunology , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Adolescent , Adult , Aged , Cell Line , Child , Child, Preschool , Female , Hepatitis B/metabolism , Hepatitis B/pathology , Hepatitis B/virology , Hepatitis B virus/genetics , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Male , Middle Aged , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Young Adult
15.
Antivir Ther ; 13(2): 221-30, 2008.
Article in English | MEDLINE | ID: mdl-18505173

ABSTRACT

BACKGROUND: Chronic infection with hepatitis B virus (HBV) is a major factor associated with the development of hepatocellular carcinoma, but the mechanism by which this occurs is unknown. Treatment of chronic hepatitis B with lamivudine results in virological suppression and histological improvement; however, the role of lamivudine in preventing the development of hepatocellular carcinoma is less well defined. We recently reported that replication of HBV in a cell-culture system was associated with the upregulation of pERK, pAkt, pc-Myc, nuclear cyclin B1, p21(cip1) and p53 together with G2 cell cycle arrest. METHODS: In order to determine whether lamivudine is able to reverse the HBV-induced changes on signal transduction and cell cycle, we infected Huh7 cells with a recombinant adeno-HBV virus in the presence of 0-50 microM of lamivudine. Signal transduction and cell cycle regulatory proteins were analysed by western immunoblot. RESULTS: Although lamivudine was able to inhibit HBV replication, it failed to reverse the changes on ERK and Akt phosphorylation. Correspondingly, levels of phospho-GSK3beta and p21(cp1/waf1) were increased, as were cyclin D1, cyclin B1, p53 and pc-Myc. CONCLUSIONS: Lamivudine was ineffective in reversing the HBV-induced changes in signal transduction pathways and cell cycle regulatory proteins, indicating that the HBV-infected cells remained primed for oncogenic transformation despite viral suppression.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B virus/pathogenicity , Lamivudine/pharmacology , Signal Transduction/drug effects , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Line , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatitis B virus/enzymology , Humans , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism
16.
Intervirology ; 51(6): 432-43, 2008.
Article in English | MEDLINE | ID: mdl-19321929

ABSTRACT

OBJECTIVE: The hepatitis B virus X (HBx) protein plays an important role in the pathogenesis of hepatocellular carcinoma (HCC). One potential mechanism by which HBx can cause liver cancer may involve intracellular distribution and consecutively modulation of the proliferative important STAT/SOCS signaling with upregulation of STAT3. METHODS: 153 Vietnamese HBV-infected patients, including 48 patients with HCC, were analyzed. HBx sequences were determined by sequencing and subcloned for functional experiments. Intracellular localization of HBx mutants was determined by immunofluorescence assays. The impact of HBx mutants on JAK/STAT/SOCS signaling was investigated using Western blot and PCR analyses. RESULTS: In 4/48 HCC patients, truncated HBx together with full-length mutated HBx proteins were observed. Expression of HBx mutant proteins demonstrated an atypical nuclear and perinuclear localization. Functional experiments to determine the effect of HBx mutants on STAT/SOCS signaling demonstrated a significantly increased upregulation of STAT3 activation (p > 0.001) in comparison to wild-type (wt)-HBx. STAT1 was not activated either by wt-HBx or HBx mutants. Interestingly, SOCS1 and SOCS3 expression was not activated by wt-HBx and HBx mutants. CONCLUSIONS: Our results suggest that atypical nuclear/perinuclear localization of HBx mutants might be responsible for an enhanced activation of STAT3, inhibition of STAT1 and silencing of SOCS1/SOCS3 expression. This observation points to an active role of HBx mutants in hepatocarcinogenesis that involves dysregulation of STAT/SOCS signaling.


Subject(s)
Hepatitis B virus , Hepatitis B, Chronic , STAT Transcription Factors/metabolism , Signal Transduction , Suppressor of Cytokine Signaling Proteins/metabolism , Trans-Activators/physiology , Adult , Aged , Amino Acid Sequence , Carcinoma, Hepatocellular/physiopathology , Carcinoma, Hepatocellular/virology , Cell Line, Tumor , Female , Gene Expression Regulation, Viral , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B, Chronic/physiopathology , Hepatitis B, Chronic/virology , Humans , Male , Middle Aged , Mutation , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Sequence Alignment , Suppressor of Cytokine Signaling 1 Protein , Suppressor of Cytokine Signaling 3 Protein , Time Factors , Trans-Activators/chemistry , Trans-Activators/genetics , Trans-Activators/metabolism , Viral Regulatory and Accessory Proteins
17.
J Clin Virol ; 39(4): 288-94, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17569577

ABSTRACT

BACKGROUND: The role of neutralizing antibody (NAb) in determining response to antiviral therapy has not been established. OBJECTIVE: In this study we have analysed the kinetic's of the NAb response in patients with chronic hepatitis C who received antiviral therapy. STUDY DESIGN: Seventeen patients infected with genotype 1, 2a/c or 3a hepatitis C virus (HCV) were enrolled, eight with a sustained virological response (SVR), five non-responders and four relapsers. RESULTS: The mean NAb titre required to neutralize 50% of the E1E2-pp in patients who achieved an SVR (294+/-S.D. 51), in relapsers (246+/-S.D. 61.7) and non-responders (286+/-S.D. 80.95) did not differ significantly between the patient groups and did not alter during the course of treatment (P>0.01). Genetic variation present before antiviral therapy was analysed by single strand conformation polymorphism (SSCP) and failed to demonstrate a significant difference in the mean number of amplified E1E2 DNA fragments from the serum of patients who achieved an SVR (3.15+/-S.D. 1.53), relapsers (2.8+/-S.D. 1.32) or non-responders (3.69+/-S.D. 1.75). The baseline serum HCV viral loads were also not significantly different between patients who achieved an SVR (1.4 x 10(6) copies/ml; +/-S.D. 2.4 x 10(6)), relapsers (1.3 x 10(7) copies/ml; +/-S.D. 2.4 x 10(7)) and non-responders (1.5 x 10(6) copies/ml; +/-S.D. 1.1 x 10(6)). CONCLUSION: We have shown that neutralizing anti-HCVpp antibody is not associated with response to antiviral therapy. In addition, there was no correlation between baseline virological load, circulating viral quasi-species, NAb titres and final response to treatment.


Subject(s)
Antiviral Agents/therapeutic use , Hepacivirus/immunology , Hepatitis C Antibodies/immunology , Hepatitis C, Chronic/drug therapy , Hepatitis C, Chronic/immunology , Drug Therapy, Combination , Hepacivirus/classification , Hepacivirus/drug effects , Hepacivirus/genetics , Hepatitis C Antibodies/blood , Hepatitis C, Chronic/virology , Humans , Interferons/therapeutic use , Neutralization Tests , Polyethylene Glycols/therapeutic use , Polymorphism, Single-Stranded Conformational , Ribavirin/therapeutic use , Viral Envelope Proteins/immunology
18.
J Hepatol ; 47(3): 325-37, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17512084

ABSTRACT

BACKGROUND/AIMS: Chronic infection with the hepatitis B virus (HBV) is strongly associated with the development of hepatocellular carcinoma but the mechanism by which this occurs is unknown. Numerous studies have focused on the HBV X protein showing that it activates signal transduction pathways while few have investigated these changes in HBV-replicating hepatocytes. METHODS: We utilized the recombinant adenovirus system to deliver a replication competent HBV genome into Huh7 and primary marmoset hepatocytes (PMH) to examine the effects of active viral replication on the regulation of Ras-ERK signal transduction and related pathways. RESULTS: Huh7 cells and PMHs replicating HBV demonstrated significant upregulation in phosphorylated ERK, Akt, c-myc together with increased p53, cyclin B1 and p21(cip1) expression and cell cycle progression to G2 phase in the absence of increased cell proliferation. Phosphorylation of the key cell survival kinase, Akt, was significantly increased, resulting in increased serine phosphorylation of the downstream target, GSK3-beta. CONCLUSIONS: These results demonstrated simultaneous activation of the MAP Kinase and Akt pathways in HBV-replicating hepatocytes that resulted in dysregulation in the control of cell cycle progression and which help explain the early pathogenic mechanisms that underlie malignant transformation associated with chronic hepatitis B infection.


Subject(s)
Carcinoma, Hepatocellular/virology , Cell Cycle , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/physiopathology , Liver Neoplasms/virology , MAP Kinase Signaling System , Virus Replication , Animals , Callithrix , Carcinoma, Hepatocellular/physiopathology , Cell Line , Cell Transformation, Neoplastic , Cyclin A/metabolism , Cyclin B/metabolism , G2 Phase , Hepatitis B, Chronic/enzymology , Hepatocytes/virology , Liver Neoplasms/physiopathology , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-myc/metabolism
19.
Rev Med Virol ; 13(4): 255-72, 2003.
Article in English | MEDLINE | ID: mdl-12820187

ABSTRACT

The clinical management of chronic hepatitis B infection has entered a new era with the introduction and widespread use of oral nucleoside analogues such as lamivudine and nucleotides such as adefovir dipivoxil. From this, new challenges have now emerged in terms of preventing antiviral drug resistance, promoting viral clearance and improving long-term survival. For example, the natural history of nucleoside or nucleotide analogue-associated hepatitis B virus resistant mutants has yet to be determined. Furthermore, the increasing prevalence of HBeAg negative disease with its reduced response to current therapies represents an ongoing challenge to attempts to improve standard of care. There is increasing recognition of the pivotal role that viral load and genotype, and their complex interactions with the host immune response, play in determining the outcome of these treatment interventions. The purpose of this paper is to highlight several key factors that should be considered in the context of future clinical research and management of chronic hepatitis B.


Subject(s)
Hepatitis B, Chronic/drug therapy , Antiviral Agents/therapeutic use , DNA, Viral/genetics , Drug Resistance, Viral/genetics , Gene Products, pol/chemistry , Gene Products, pol/genetics , Gene Products, pol/physiology , Genes, Viral , Hepatitis B virus/drug effects , Hepatitis B virus/genetics , Hepatitis B virus/physiology , Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Humans , Mutation , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL
...