Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Viral Hepat ; 20(4): e56-65, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23490390

ABSTRACT

DNA-based vaccination appears of promise for chronic hepatitis B immunotherapy, although there is an urgent need to increase its efficacy. In this preclinical study, we evaluated the therapeutic benefit of cytokine (IL-2, IFN-γ) genes co-delivery with DNA vaccine targeting hepadnaviral proteins in the chronic duck hepatitis B virus (DHBV) infection model. Then, we investigated the persistence of replication-competent virus in the livers of apparently resolved animals. DHBV carriers received four injections of plasmids encoding DHBV envelope and core alone or co-delivered with duck IL-2 (DuIL-2) or duck IFN-γ (DuIFN-γ) plasmids. After long-term (8 months) follow-up, viral covalently closed circular (ccc) DNA was analysed in duck necropsy liver samples. Liver homogenates were also tested for in vivo infectivity in neonatal ducklings. Co-delivery of DuIFN-γ resulted in significantly lower mean viremia starting from week 21. Viral cccDNA was undetectable by conventional methods in the livers of 25% and 57% of animals co-immunized with DuIL-2 and DuIFN-γ, respectively. Interestingly, inoculation of liver homogenates from 7 such apparently resolved animals, exhibiting cccDNA undetectable in Southern blotting and DHBV expression undetectable or restricted to few hepatocytes, revealed that three liver homogenates transmitted high-titre viremia (3-5×10(10) vge/mL) to naïve animals. In conclusion, our results indicate that IFN-γ gene co-delivery considerably enhances immunotherapeutic efficacy of DNA vaccine targeting hepadnaviral proteins. Importantly, we also showed that livers exhibiting only minute amounts of hepadnaviral cccDNA could induce extremely high-titre infection, highlighting the caution that should be taken in occult hepatitis B patients to prevent HBV transmission in liver transplantation context.


Subject(s)
Hepadnaviridae Infections/therapy , Hepatitis B Vaccines/immunology , Hepatitis B Virus, Duck/immunology , Hepatitis, Viral, Animal/therapy , Interferon-gamma/immunology , Interleukin-2/immunology , Vaccines, DNA/immunology , Animals , Carrier State/therapy , Carrier State/virology , DNA, Viral/isolation & purification , Ducks , Follow-Up Studies , Hepadnaviridae Infections/virology , Hepatitis B Vaccines/administration & dosage , Hepatitis B Vaccines/genetics , Hepatitis B Virus, Duck/genetics , Hepatitis, Viral, Animal/virology , Interferon-gamma/administration & dosage , Interferon-gamma/genetics , Interleukin-2/administration & dosage , Interleukin-2/genetics , Liver/virology , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics , Viral Load , Viremia/therapy , Viremia/virology
2.
Antimicrob Agents Chemother ; 48(7): 2683-92, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15215126

ABSTRACT

Our aim was to evaluate the antiviral effect of a combination of two nucleoside reverse transcriptase inhibitors, emtricitabine (FTC) and clevudine (L-FMAU), with the addition of an adenovirus-driven delivery of recombinant gamma interferon (IFN-gamma) in the woodchuck model of hepatitis B virus infection. Six woodchuck hepatitis virus (WHV)-infected woodchucks received L-FMAU (10 mg/kg) plus FTC (30 mg/kg) intraperitoneally for 8 weeks; six other animals received in addition an intravenous injection of a recombinant adenovirus vector expressing woodchuck IFN-gamma (Ad-IFN) at weeks 4 and 8. In the control group, two animals received Ad-IFN alone, two received adenovirus vector expressing the green fluorescent protein reporter gene, and one remained untreated. In less than 2 weeks, all woodchucks that received L-FMAU plus FTC showed a rapid and marked inhibition of viral replication, with a 4-log(10) drop in serum WHV DNA. In two animals, viremia remained suppressed for several months after the end of treatment. Similarly, a dramatic decrease in intrahepatic replicative intermediates of viral DNA was observed in the L-FMAU/FTC-treated groups. The additional administration of Ad-IFN led to increased inflammation in the liver but did not enhance the antiviral effect of the L-FMAU/FTC combination. In conclusion, therapies combining L-FMAU and FTC in WHV-infected woodchucks resulted in a potent and sustained antihepadnaviral effect both in the liver and in the blood circulation. However, no extra benefit of adding IFN-gamma gene transduction to the L-FMAU/FTC combination could be detected.


Subject(s)
Adenoviridae/genetics , Antiviral Agents/therapeutic use , Arabinofuranosyluracil/analogs & derivatives , Arabinofuranosyluracil/therapeutic use , Deoxycytidine/analogs & derivatives , Deoxycytidine/therapeutic use , Genetic Therapy , Hepatitis B Virus, Woodchuck , Hepatitis B/therapy , Interferon-gamma/genetics , Interferon-gamma/therapeutic use , Marmota/physiology , Reverse Transcriptase Inhibitors/therapeutic use , Animals , DNA-Directed DNA Polymerase/metabolism , Drug Combinations , Emtricitabine , Hepatitis B/drug therapy , Hepatitis B Core Antigens/metabolism , Liver/metabolism , Microscopy, Confocal , Microscopy, Electron , Mitochondria, Liver/enzymology , Proliferating Cell Nuclear Antigen/metabolism , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction , Viremia/virology , Virus Replication/drug effects , Virus Replication/physiology
3.
Cancer Detect Prev ; 25(2): 192-201, 2001.
Article in English | MEDLINE | ID: mdl-11341355

ABSTRACT

The aim of our study was to use the Pekin duck model to investigate the interactions between hepadnaviral infection and aflatoxin B1 (AFB1) exposure including the role of both factors in the induction of oxidative stress in the liver. AFB1 exposure of duck hepatitis B virus (DHBV) infected Pekin ducks induced a significant increase in viral replication associated with an intense biliary ductular cells proliferation. Interestingly, extremely high levels of AFB1-DNA adducts (40-120 pmol AFB1-Fapy/mg DNA) and AFB1-albumin adducts (1,500-3,000 pg AFB1-lys Eq/mg albumin) were detected in duck liver and serum respectively, as compared to other animal species exposed to a similar AFB1 dose. DHBV infection was found to induce a non-significant increase in AFB1-albumin adduct levels in duck serum. During the treatment duration there was no effect on formation of oxidative base damage within DNA and no effect on oxidative lipid peroxidation following either viral infection or AFB1 exposure. In terms of hepatic antioxidant enzymes (catalase, superoxide dismutase (SOD), glutathione peroxidase) a significant increase in SOD activity occurred following AFB1 exposure, but not DHBV infection, but this was observed only after the cessation of treatment, when biliary ductular cells proliferation was reduced.


Subject(s)
Aflatoxin B1/toxicity , Hepadnaviridae Infections/metabolism , Hepatitis B Virus, Duck/physiology , Hepatitis, Viral, Animal/metabolism , Liver/drug effects , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Animals , Catalase/metabolism , Cattle , DNA/metabolism , DNA Adducts/metabolism , DNA, Viral/blood , DNA, Viral/metabolism , Deoxycytidine/analogs & derivatives , Deoxycytidine/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Ducks , Glutathione Peroxidase/metabolism , Hepadnaviridae Infections/virology , Hepatitis, Viral, Animal/virology , Lipid Peroxidation , Serum Albumin, Bovine/metabolism , Superoxide Dismutase/metabolism
4.
Antimicrob Agents Chemother ; 45(4): 1065-77, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11257017

ABSTRACT

The L-nucleoside analog beta-L-2',3'-dideoxy-2',3'-didehydro-5-fluorocytidine (beta-L-Fd4C) was first shown to exhibit potent activity against hepatitis B virus (HBV) in tissue culture and then to significantly inhibit viral spread during acute infection in the duck HBV model (F. Le Guerhier et al., Antimicrob. Agents Chemother. 44:111-122, 2000). We have therefore examined its antiviral activity in a mammalian model of chronic HBV infection, the woodchuck chronically infected with woodchuck hepatitis virus (WHV). Side-by-side comparison of beta-L-Fd4C and lamivudine administered intraperitoneally during short-term and long-term protocols demonstrated a more profound inhibition of viremia in beta-L-Fd4C-treated groups. Moreover, beta-L-Fd4C induced a marked inhibition of intrahepatic viral DNA synthesis compared with that induced by lamivudine. Nevertheless, covalently closed circular (CCC) DNA persistence explained the lack of clearance of infected hepatocytes expressing viral antigens and the relapse of WHV replication after drug withdrawal. Liver histology showed a decrease in the inflammatory activity of chronic hepatitis in woodchucks receiving beta-L-Fd4C. An electron microscopy study showed the absence of ultrastructural changes of hepatic mitochondria, biliary canaliculi, and bile ducts. However, a loss of weight was observed in all animals, whatever the treatment, as was a transient skin pigmentation in all woodchucks during beta-L-Fd4C treatment. There was no evidence that lamivudine or beta-L-Fd4C could prevent the development of hepatocellular carcinoma with the protocols used. These results indicate that beta-L-Fd4C exhibits a more potent antiviral effect than lamivudine in the WHV model but was not able to eradicate CCC DNA and infected cells from the liver at the dosage and with the protocol used.


Subject(s)
Antiviral Agents/therapeutic use , Hepatitis B Virus, Woodchuck , Hepatitis B, Chronic/drug therapy , Zalcitabine/therapeutic use , Animals , Antiviral Agents/administration & dosage , Antiviral Agents/adverse effects , Carcinoma, Hepatocellular/prevention & control , DNA, Circular/drug effects , DNA, Viral/drug effects , Drug Administration Schedule , Hepatitis B Virus, Woodchuck/genetics , Hepatitis B, Chronic/pathology , Hepatitis B, Chronic/virology , Lamivudine/therapeutic use , Liver/pathology , Liver/ultrastructure , Liver/virology , Marmota , Skin Pigmentation/drug effects , Viremia/drug therapy , Viremia/pathology , Viremia/virology , Virus Replication/drug effects , Zalcitabine/administration & dosage , Zalcitabine/adverse effects , Zalcitabine/analogs & derivatives
5.
J Hepatol ; 34(1): 114-22, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11211887

ABSTRACT

BACKGROUND/AIMS: Hepatitis B virus mutants of the polymerase gene are frequently selected during lamivudine therapy for chronic hepatitis B. To study the biology of these mutants, we analyzed their replication capacity in the duck hepatitis B virus (DHBV) infection. METHODS: The B and C domain polymerase mutants corresponding to the clinical isolates were engineered by site directed mutagenesis in the DHBV genome in different expression vectors. RESULTS: The study of the enzymatic activity of the mutated viral polymerase polypeptides analyzed in a cell free system demonstrated a lower priming activity and a decreased capacity of elongation of viral minus strand DNA that was consistent with the lower replication capacity of these mutants in transfected leghorn male hepatoma cells compared to wild type genome. These mutants had a lower replication capacity in primary hepatocytes and in in vivo transfected ducklings. Although resistant to lamivudine, these mutants remained sensitive to PMEA. CONCLUSION: YMDD mutants of the DHBV reverse transcriptase have a decreased replication capacity both in vitro and in vivo, and are not cross-resistant to PMEA. These results may be important to design new antiviral strategies to combat the replication of the lamivudine resistant viral strains.


Subject(s)
Antiviral Agents/pharmacology , Hepatitis B Virus, Duck/enzymology , Lamivudine/pharmacology , Mutation , Organophosphonates , RNA-Directed DNA Polymerase/genetics , Adenine/analogs & derivatives , Adenine/pharmacology , Amino Acid Sequence , DNA, Viral/biosynthesis , Drug Resistance , Hepatitis B Virus, Duck/drug effects , Hepatitis B Virus, Duck/genetics , Molecular Sequence Data , RNA-Directed DNA Polymerase/physiology , Transcription, Genetic
6.
Vaccine ; 18(27): 3091-6, 2000 Jul 15.
Article in English | MEDLINE | ID: mdl-10856788

ABSTRACT

DNA vaccination may represent an interesting strategy for early life immunization. However, in some cases, this approach has been shown to induce a tolerance rather than immunity. We have compared the efficiency of neonatal DNA or protein immunization against hepadnavirus envelope protein using the duck hepatitis B virus (DHBV) model. Three-day-old ducklings were immunized with either a plasmid encoding the DHBV pre-S/S large envelope protein (L), or a recombinant preS protein, followed by sequential DNA or protein boosts at weeks 4 and 15. Our results showed that genetic immunization of duck neonates induced specific humoral response to DHBV L protein. Interestingly, an enhanced antibody response was elicited when animals received DNA priming-DNA boosting as compared to DNA priming-protein boosting.


Subject(s)
Animals, Newborn/immunology , Hepatitis Antibodies/blood , Hepatitis B Virus, Duck/immunology , Vaccines, DNA/immunology , Viral Envelope Proteins/immunology , Viral Vaccines/immunology , Animals , Ducks , Immunization , Immunization Schedule
7.
J Virol ; 74(10): 4908-11, 2000 May.
Article in English | MEDLINE | ID: mdl-10775634

ABSTRACT

The outcome and protective efficacy of maternal antibodies elicited by DNA immunization to the large (L) hepadnavirus envelope protein were studied using the duck hepatitis B virus (DHBV) model. Following genetic immunization of breeding ducks with a DHBV L protein gene-bearing plasmid, specific and highly neutralizing antibodies were transferred from the sera of immunized ducks, via the egg yolk, to the progeny of vaccinees. Interestingly, large amounts (60 to 100 mg/egg) of high-titer and L protein-specific yolk immunoglobulins (immunoglobulin Y) accumulated in the egg yolk. These results suggest that eggs from genetically immunized avians may represent a potent source of DNA-designed antibodies specific to viral antigen. Importantly, these antibodies are vertically transmitted and protect offspring against high-titer DHBV challenge.


Subject(s)
Hepadnaviridae Infections/prevention & control , Hepatitis Antibodies/immunology , Hepatitis B Virus, Duck/immunology , Immunity, Maternally-Acquired , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Ducks , Hepadnaviridae Infections/immunology , Hepatitis B Virus, Duck/metabolism , Vaccination , Vaccines, DNA/administration & dosage , Viral Envelope Proteins/administration & dosage , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology , Viral Vaccines/administration & dosage , Viral Vaccines/immunology
8.
Antimicrob Agents Chemother ; 44(1): 111-22, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10602731

ABSTRACT

A novel L-nucleoside analog of deoxycytidine, 2',3'-dideoxy-2', 3'-didehydro-beta-L-5-fluorocytidine (beta-L-Fd4C), was recently shown to strongly inhibit hepatitis B virus (HBV) replication in the 2.2.15 cell line. Therefore, its antiviral activity was evaluated in the duck HBV (DHBV) infection model. Using a cell-free system for the expression of the DHBV polymerase, beta-L-Fd4C-TP exhibited a concentration-dependent inhibition of dCTP incorporation into viral minus-strand DNA with a 50% inhibitory concentration of 0.2 microM which was lower than that of other tested deoxycytidine analogs, i.e. , lamivudine-TP, ddC-TP, and beta-L-FddC-TP. Further analysis showed that beta-L-Fd4C-TP is likely to be a competitive inhibitor of dCTP incorporation and to cause premature DNA chain termination. In primary duck hepatocyte cultures infected in vitro, beta-L-Fd4C administration exhibited a long-lasting inhibitory effect on viral DNA synthesis but could not clear viral covalently closed circular DNA (CCC DNA). Results of short-term antiviral treatment in experimentally infected ducklings showed that beta-L-Fd4C exhibited the most potent antiviral effect, followed by beta-L-FddC, lamivudine, and ddC. Longer administration of beta-L-Fd4C induced a sustained suppression of viremia (>95% of controls) and of viral DNA synthesis within the liver. However, the persistence of trace amounts of viral CCC DNA detected only by PCR was associated with a recurrence of viral replication after drug withdrawal. In parallel, beta-L-Fd4C treatment suppressed viral antigen expression within the liver and decreased intrahepatic inflammation and was not associated with any sign of toxicity. Our data, therefore, demonstrate that in the duck model of HBV infection, beta-L-Fd4C is a potent inhibitor of DHBV reverse transcriptase activity in vitro and suppresses viral replication in the liver in vivo.


Subject(s)
Antiviral Agents/therapeutic use , Hepadnaviridae Infections/drug therapy , Hepatitis B Virus, Duck , Zalcitabine/analogs & derivatives , Animals , DNA, Circular/biosynthesis , DNA, Viral/biosynthesis , Ducks , Hepadnaviridae Infections/pathology , Liver/pathology , Reverse Transcriptase Inhibitors/therapeutic use , Viral Proteins/biosynthesis , Virus Replication/drug effects , Zalcitabine/metabolism , Zalcitabine/therapeutic use
9.
Hepatology ; 29(4): 1317-23, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10094981

ABSTRACT

Epidemiological studies have suggested synergistic interactions between chronic hepatitis B virus (HBV) infection and aflatoxin B1 (AFB1) exposure in the etiology of hepatocellular carcinoma (HCC), although the molecular mechanisms of their interactions are still not understood. The aim of this study was to use the Pekin duck model to investigate the impact of AFB1 exposure on duck hepatitis B virus (DHBV) replication during the early stages of virus-carcinogen interactions. Six-week-old chronic DHBV-carrier or uninfected ducks were exposed to AFB1 for 5 weeks or treated with dimethylsulfoxide (DMSO) as a control. Animals were observed for 6 to 13 weeks after AFB1 treatment to study the influence of AFB1 exposure on DHBV replication and liver pathologies. Histological analysis showed more marked changes in the livers of AFB1-treated ducks, and these were enhanced by DHBV infection. A significant increase in serum and liver DHBV DNA level was observed in AFB1-treated ducks as compared with DMSO-treated controls. In addition, viral RNAs, in particular the pregenomic RNA that is the template of viral replication, and intrahepatic DHBV DNA replicative intermediates, were significantly increased by AFB1 treatment. Moreover, an overexpression and accumulation of DHBV large envelope (L) protein was observed in the hepatocytes of AFB1-exposed animals. The in vitro study has further confirmed an increase in intracellular viral DNA and in virus release in AFB1-treated primary duck hepatocytes. Taken together, our results indicate that AFB1 exposure leads to an increase in virus gene expression associated with intrahepatic accumulation of DHBV L protein and enhanced liver pathology.


Subject(s)
Aflatoxin B1/pharmacology , Carcinogens/pharmacology , Gene Expression Regulation, Viral/drug effects , Hepatitis B Virus, Duck/genetics , Animals , Animals, Newborn , Cells, Cultured , DNA, Viral/blood , DNA, Viral/metabolism , Dose-Response Relationship, Drug , Ducks , Hepatitis B Virus, Duck/metabolism , Liver/drug effects , Liver/pathology , Liver/virology , Male , RNA, Viral/metabolism , Viral Envelope Proteins/metabolism , Virus Replication/drug effects
10.
Antiviral Res ; 40(3): 179-87, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10027652

ABSTRACT

The in vitro and in vivo antiviral activities of two mononucleoside phosphotriester derivatives of acyclovir (ACV) incorporating S-acyl-2-thioethyl (SATE) groups are reported using the duck model of hepatitis B (DHBV). In primary duck hepatocyte cultures, the described phosphotriesters significantly inhibited the replication of DHBV at submicromolar concentrations. They were found to be more potent than the parent nucleoside. This result was in agreement with our data concerning the anti-HBV activity of these pronucleotides in HepG2.2.15 cells (previous paper). In vivo, the studied SATE pronucleotide was also found to be more efficient than ACV in infected ducklings upon short-term oral therapy, while intraperitoneal treatment showed high anti-DHBV activity with both ACV and its SATE pronucleotide in this animal model. These findings demonstrate the potential of SATE pronucleotides of ACV as anti-HBV agents.


Subject(s)
Acyclovir/analogs & derivatives , Acyclovir/pharmacology , Antiviral Agents/pharmacology , Hepadnaviridae Infections/drug therapy , Hepatitis B Virus, Duck/drug effects , Organophosphonates/pharmacology , Acyclovir/therapeutic use , Animals , Antiviral Agents/therapeutic use , Blotting, Southern , Cells, Cultured , DNA, Viral , Drug Evaluation, Preclinical , Ducks , Hepadnaviridae Infections/virology , Hepatitis B Virus, Duck/physiology , Liver/cytology , Liver/virology , Nucleotides/chemical synthesis , Nucleotides/metabolism , Organophosphonates/therapeutic use , Prodrugs/chemical synthesis , Prodrugs/metabolism , Virus Replication/drug effects
11.
Gastroenterology ; 116(3): 658-65, 1999 Mar.
Article in English | MEDLINE | ID: mdl-10029625

ABSTRACT

BACKGROUND & AIMS: Studies in the murine model suggest that injection of DNA encoding hepatitis B virus structural proteins is promising for the induction of a specific immune response. We used the duck hepatitis B virus (DHBV) model to study the protective and therapeutic effects of naked DNA immunization against hepadnaviral large envelope protein. METHODS: A pCI-preS/S plasmid expressing the DHBV large protein was used for intramuscular immunization of ducks. The humoral response was tested by enzyme-linked immunosorbent assay, immunoblotting, neutralization, and in vivo protection tests. For DNA therapy, DHBV-carrier ducks received four injections of this plasmid. Viremia was monitored for 10 months; thereafter, liver biopsies were performed. RESULTS: Immunization with pCI-preS/S plasmid induced a specific, long-lasting, neutralizing, and highly protective anti-preS humoral response in uninfected animals. After pCI-preS/S treatment, a significant and sustained decrease in serum and liver DHBV DNA was observed for carrier ducks compared with the controls. CONCLUSIONS: DNA immunization against DHBV large protein results in a potent and protective anti-preS response in the duck model. The results of long-term follow-up of DNA-treated chronically infected ducks are promising and show the usefulness of this model for the study of genetic immunization in chronic hepatitis B therapy.


Subject(s)
Hepadnaviridae Infections/veterinary , Hepadnaviridae/immunology , Hepatitis B Virus, Duck/immunology , Poultry Diseases/immunology , Vaccines, Synthetic , Viral Envelope Proteins/immunology , Viral Vaccines , Animals , Antibody Formation , Bupivacaine/therapeutic use , Carrier State/immunology , Carrier State/veterinary , Ducks , Enzyme-Linked Immunosorbent Assay , Hepadnaviridae Infections/immunology , Hepadnaviridae Infections/prevention & control , Hepatitis B Antibodies/blood , Plasmids , Poultry Diseases/prevention & control
12.
Antimicrob Agents Chemother ; 42(2): 369-76, 1998 Feb.
Article in English | MEDLINE | ID: mdl-9527788

ABSTRACT

The antiviral activity of 2'-fluoro-5-methyl-beta-L-arabinofuranosyluracil (L-FMAU), a novel L-nucleoside analog of thymidine known to be an inhibitor of hepatitis B virus (HBV) replication in hepatoma cells (2.2.1.5 cell line), was evaluated in the duck HBV (DHBV) model. Short-term oral administration (5 days) of L-FMAU (40 mg/kg of body weight/day) to experimentally infected ducklings induced a significant decrease in the level of viremia. This antiviral effect was sustained in animals when therapy was prolonged for 8 days. The histological study showed no evidence of liver toxicity in the L-FMAU-treated group. By contrast, microvesicular steatosis was found in the livers of dideoxycytidine-treated animals. L-FMAU administration in primary duck hepatocyte cultures infected with DHBV induced a dose-dependent inhibition of both virion release in culture supernatants and intracellular viral DNA synthesis, without clearance of viral covalently closed circular DNA. By using a cell-free system for the expression of an enzymatically active DHBV reverse transcriptase, it was shown that L-FMAU triphosphate exhibits an inhibitory effect on the incorporation of dAMP in the viral DNA primer. Thus, our data demonstrate that L-FMAU inhibits DHBV replication in vitro and in vivo. Long-term administration of L-FMAU for the eradication of viral infection in animal models of HBV infection should be evaluated.


Subject(s)
Antiviral Agents/pharmacology , Arabinofuranosyluracil/analogs & derivatives , DNA, Viral/drug effects , Hepadnaviridae Infections/drug therapy , Hepatitis B Virus, Duck/drug effects , Virus Replication/drug effects , Administration, Oral , Animals , Antiviral Agents/administration & dosage , Arabinofuranosyluracil/administration & dosage , Arabinofuranosyluracil/pharmacology , DNA, Viral/metabolism , Ducks , Hepadnaviridae Infections/virology , Hepatitis B Virus, Duck/physiology , Liver/drug effects , Liver/pathology
13.
Virology ; 234(2): 291-9, 1997 Aug 04.
Article in English | MEDLINE | ID: mdl-9268160

ABSTRACT

To better understand the role of specific residues within the duck hepatitis B virus (DHBV) pre-S protein in neutralization and infectivity, we have selected and identified pre-S variants which escape neutralization. A highly neutralizing monoclonal antibody (Mab 900) which recognizes an epitope 83IPQPQWTP90 localized previously on the DHBV pre-S protein, within a region suspected to mediate the virus interaction with hepatocytes, was used as immune pressure. After only two in vivo neutralization rounds with Mab 900, five different pre-S mutant genomes were identified, which harbored point mutations affecting only proline residues located at position 90 within this epitope (83IPQPQWTP90) and/or at a distance at position 5. We have shown that a single (P5L) or double proline (P5L + P90H) substitution affect neither virus replication capacity nor in vivo infectivity. However, the P5 mutation reduces mutant recognition by Mab 900 twofold, while the substitution of both prolines 5 and 90 almost completely abolishes mutant P5L + P90H reactivity with this Mab and leads to a decrease of neutralization. Therefore we describe here an experimental system which allows rapid in vivo selection and identification of DHBV pre-S variants and provide evidence that residues within and at a distance from the neutralization epitope are important in DHBV neutralization but do not affect its replication capacity and infectivity.


Subject(s)
Antibodies, Viral/immunology , Antigens, Viral/immunology , Hepadnaviridae Infections/virology , Hepatitis B Virus, Duck/immunology , Viral Envelope Proteins , Viral Proteins/physiology , Animals , Ducks , Epitope Mapping , Epitopes/immunology , Point Mutation , Rabbits , Virus Replication
14.
J Hepatol ; 26(6): 1324-30, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9210620

ABSTRACT

BACKGROUND: Eastern American woodchuck (Marmota monax), naturally infected with woodchuck hepatitis virus, a virus similar to human hepatitis B virus, develops liver cancer with a high prevalence. AIMS: The aim of this work was to assess Marmota monax as a model of human hepatocellular carcinoma, especially to assess new potential adjuvant therapies after surgical resection. METHODS: Forty-four woodchuck hepatitis virus-infected animals were regularly screened by ultrasound examination from the age of 18 months and for a 30-month period. One or more liver tumors were diagnosed in 31 animals (70%). Five of them with multifocal tumor or poor general status were considered unsuitable for surgery. The other 26 were operated on. At laparotomy no tumor was found in three. RESULTS: The 18 liver tumors studied were hepatocellular carcinomas, grossly and microscopically similar to human hepatocellular carcinoma. Peritumoral parenchyma studied in 13 specimens was always non-cirrhotic but adequate staining demonstrated patterns of fibrosis in four cases. Clear evidence of chronic active hepatitis, periportal hepatitis and steatosis were demonstrated in five, seven and one of the 13 specimens, respectively. Tumors were treated by tumorectomy in eight animals, by alcoholization in seven and by laser photocoagulation in one. A simple tumor biopsy was performed in the other seven. Ten animals died postoperatively. All the survivors in the tumorectomy group died from tumor recurrence within 10-18 months after surgery. CONCLUSIONS: It is concluded that woodchuck hepatitis virus-induced liver carcinoma is a natural model of human hepatocellular carcinoma with similar pathology and natural history, including early ultrasonic detection and tumor recurrence after resection. Tumor excision is feasible in this animal model, which now provides the basis for assessment of new potential adjuvant therapies for human hepatocellular carcinoma in an attempt to reduce the high recurrence rate after surgical resection in humans.


Subject(s)
Carcinoma, Hepatocellular/surgery , Carcinoma, Hepatocellular/veterinary , Hepatitis B Virus, Woodchuck , Hepatitis B/veterinary , Liver Neoplasms/surgery , Liver Neoplasms/veterinary , Animals , Carcinoma, Hepatocellular/diagnostic imaging , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Hepatitis B/pathology , Humans , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/pathology , Marmota , Ultrasonography
15.
Parasite ; 3(2): 175-8, 1996 Jun.
Article in French | MEDLINE | ID: mdl-8758553

ABSTRACT

Autopsy of two Woodchuck Hepatitis Virus (WHV) infected Woodchucks, Marmota monax, revealed the presence of two parasites in an hepatic localization, Taenia mustelae (Larvae) and Calodium hepaticum. The authors present the identification of the two parasites, based on the observation of cysticerci of Taenia mustelae, or on the observation of the eggs of C. hepaticum. They discuss the probable interaction between hepatic parasites and WHV infection.


Subject(s)
Helminthiasis, Animal , Hepatitis B Virus, Woodchuck , Hepatitis B/veterinary , Liver Diseases, Parasitic/veterinary , Marmota/parasitology , Marmota/virology , Animals , Helminthiasis/complications , Hepatitis B/complications , Liver/parasitology , Liver/pathology , Liver Diseases, Parasitic/complications
16.
J Virol ; 70(5): 2691-6, 1996 May.
Article in English | MEDLINE | ID: mdl-8627742

ABSTRACT

During chronic hepadnavirus infection, virus persistence depends on the regulation of the pool of covalently closed circular DNA (cccDNA), which is the template for transcription of viral RNA species. The development of in vitro infection of duck hepatocyte primary cultures by duck hepatitis B virus (DHBV) provides a unique opportunity to study the regulation of cccDNA synthesis. After DHBV in vitro infection, cccDNA is detected 1 day later and is amplified to a high copy number after 1 week in culture. We studied whether this amplification occurs during cell cycle progression of duckling hepatocytes. By using [3H]thymidine incorporation, we found that hepatocytes obtained from 3-week-old ducklings spontaneously entered the S phase of the cell cycle when cultured in serum-free medium without added growth factors. Bromodeoxyuridine labeling confirmed that cellular DNA synthesis took place in more than 50% of parenchymal cells. Cytofluorometry analysis revealed the presence of asynchronous populations and polyploidization processes. The addition of a cell cycle blocker, n-butyrate, completely inhibited [3H]thymidine incorporation and blocked duckling hepatocytes in the G1 phase of the cell cycle. Simultaneously, butyrate inhibited cccDNA amplification and allowed the establishment of DHBV infection, as demonstrated by the detection of a basal level of cccDNA in treated hepatocytes. Both effects were reversible since active cell DNA synthesis was restored and cccDNA accumulated after drug withdrawal.


Subject(s)
Butyrates/pharmacology , Cell Cycle/drug effects , DNA, Circular/drug effects , Hepatitis B Virus, Duck/physiology , Liver/cytology , Animals , Blotting, Southern , Bromodeoxyuridine , Butyric Acid , Cells, Cultured , DNA, Circular/metabolism , DNA, Viral/drug effects , DNA, Viral/metabolism , Ducks , Hepatitis B Virus, Duck/drug effects , Hepatitis B Virus, Duck/genetics , Liver/drug effects , Liver/virology , RNA, Viral/biosynthesis , Templates, Genetic , Thymidine/metabolism , Transcription, Genetic , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...